Germ Cell Development Methods And Protocols 1st Edition Marco Barchi
Germ Cell Development Methods And Protocols 1st Edition Marco Barchi
Germ Cell Development Methods And Protocols 1st Edition Marco Barchi
Germ Cell Development Methods And Protocols 1st Edition Marco Barchi
1. Germ Cell Development Methods And Protocols 1st
Edition Marco Barchi download
https://ebookbell.com/product/germ-cell-development-methods-and-
protocols-1st-edition-marco-barchi-55541864
Explore and download more ebooks at ebookbell.com
2. Here are some recommended products that we believe you will be
interested in. You can click the link to download.
Germ Cell Development Methods And Protocols Methods In Molecular
Biology 2770 1st Edition Marco Barchi
https://ebookbell.com/product/germ-cell-development-methods-and-
protocols-methods-in-molecular-biology-2770-1st-edition-marco-
barchi-55541862
Germ Cell Development In C Elegans 1st Edition Nanette Pazdernik
https://ebookbell.com/product/germ-cell-development-in-c-elegans-1st-
edition-nanette-pazdernik-4290032
Germ Cell Protocols Volume 1 Sperm And Oocyte Analysis 1st Edition
Franz Lahnsteiner Auth
https://ebookbell.com/product/germ-cell-protocols-volume-1-sperm-and-
oocyte-analysis-1st-edition-franz-lahnsteiner-auth-2330682
Germ Cell Protocols Vol 2 Molecular Embryo Analysis Live Imaging
Transgenesis And Cloning Methods In Molecular Biology Vol 254
Softcover Reprint Of Hardcover 1st Ed 2004 Heide Schatten
https://ebookbell.com/product/germ-cell-protocols-vol-2-molecular-
embryo-analysis-live-imaging-transgenesis-and-cloning-methods-in-
molecular-biology-vol-254-softcover-reprint-of-hardcover-1st-
ed-2004-heide-schatten-2418828
3. Germ Cell Tumor Angabin Matin
https://ebookbell.com/product/germ-cell-tumor-angabin-matin-2559098
Germ Cell Tumors Derek Raghavan
https://ebookbell.com/product/germ-cell-tumors-derek-raghavan-4118220
Germ Cell Tumours V The Proceedings Of The Fifth Germ Cell Tumour
Conference Devonshire Hall University Of Leeds 13th15th September 2001
1st Edition E A Rapley
https://ebookbell.com/product/germ-cell-tumours-v-the-proceedings-of-
the-fifth-germ-cell-tumour-conference-devonshire-hall-university-of-
leeds-13th15th-september-2001-1st-edition-e-a-rapley-4492024
Germ Cell Protocols Volume 1 Sperm And Oocyte Analysis 1st Edition
Franz Lahnsteiner Auth
https://ebookbell.com/product/germ-cell-protocols-volume-1-sperm-and-
oocyte-analysis-1st-edition-franz-lahnsteiner-auth-929876
Germ Cell Protocols Volume 2 Molecular Embryo Analysis Live Imaging
Transgenesis And Cloning 1st Edition Michele Boiani
https://ebookbell.com/product/germ-cell-protocols-volume-2-molecular-
embryo-analysis-live-imaging-transgenesis-and-cloning-1st-edition-
michele-boiani-929878
6. M E T H O D S I N M O L E C U L A R B I O L O G Y
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, UK
For further volumes:
http://www.springer.com/series/7651
7. For over 35 years, biological scientists have come to rely on the research protocols and
methodologies in the critically acclaimed Methods in Molecular Biology series. The series was
the first to introduce the step-by-step protocols approach that has become the standard in all
biomedical protocol publishing. Each protocol is provided in readily-reproducible step-by-
step fashion, opening with an introductory overview, a list of the materials and reagents
needed to complete the experiment, and followed by a detailed procedure that is supported
with a helpful notes section offering tips and tricks of the trade as well as troubleshooting
advice. These hallmark features were introduced by series editor Dr. John Walker and
constitute the key ingredient in each and every volume of the Methods in Molecular Biology
series. Tested and trusted, comprehensive and reliable, all protocols from the series are
indexed in PubMed.
8. Germ Cell Development
Methods and Protocols
Edited by
Marco Barchi
Department of Biomedicine and Prevention, Section of Human Anatomy, Faculty of Medicine
and Surgery, “Tor Vergata“ University of Rome, Rome, Italy
Massimo De Felici
Department of Biomedicine and Prevention, Section of Histology and Embryology, Faculty of
Medicine and Surgery, “Tor Vergata” University of Rome, Rome, Italy
10. Preface
In sexually reproducing organisms, germ cells (GSs) bear two major responsibilities: the
maintenance of the species through the transmission of hereditary traits, and species evolu-
tion through the reshuffling of the genome. During early embryonal development, GCs
develop from staminal GC precursors that give rise to primordial GCs (PGCs). The latter, by
replicating and migrating, reach the primitive gonads where they will develop into male or
female gamete precursors (spermatogonia or oogonia, respectively) depending on the
assortment of the sex chromosomes complement (XY or XX, respectively) of the somatic
gonadal cells. To reconstitute the diploid chromosome number at each fertilization, GCs
undergo meiosis. The functional role of meiosis is to reduce the genome complement by
half, which is accomplished by sequentially executing one round of DNA replication
followed by two rounds of chromosome segregation. Between DNA replication and the
first meiotic division in most organisms, paternal and maternal (homologous) chromosomes
undergo homologous recombination. The latter is key for the reshuffling of the genome and
proper segregation of homologous chromosomes at anaphase I. Importantly, recombina-
tion is initiated by a programmed wave of double strand breaks (DSBs) that occurs along
chromosomes in specific regions called “hotspots.” Precise positioning of hotspots is
essential for successful recombination; therefore, alteration of the mechanisms determining
hotspots may cause defects in pairing and synapsis of the homologous chromosomes, which
may lead to sterility or to unbalanced segregation of chromosomes at anaphase I. In the
latter case, when it is compatible with life (mostly it is the case of sex chromosomes
missegregation), it causes in humans the onset of syndromes, such as Klinefelter (47,
XXY), in which genetic-driven phenotypic manifestations are being studied.
Thus, GCs are unique in their ability to transfer genetic information across generations.
As such, proper understanding of the mechanisms underpinning their origin, regulation,
and differentiation is key to understanding alterations of these processes, which are para-
mount to the health of organisms and the survival of species. Until recent years, germ cell
research was limited by the lack of in vitro models recapitulating male or female germ cell
development. However, more recently, research in the field of stem cell biology has allowed
an impressive acceleration in the expansion of new techniques for in vitro reconstruction of
spermatogenesis and oogenesis, both in animal models and humans. In addition, the
development of somatic cell reprogramming techniques makes it possible today to obtain
stem cells from patient-derived somatic cells, providing a tool for molecular studies of
human diseases, including the syndromes resulting from defects of developing germ cells.
These advances promise to provide new insight into the understanding of basic
biological aspects of germ cell biology, as well as the opportunity for in vitro manipulation
of germ cells for toxicology studies and for genetic intervention where ethically appropriate.
In this volume, we have collected well-established protocols for the isolation, purifica-
tion, and establishment of in vitro GC systems at different stages of development in different
organisms, including chickens, mice, rats, and humans. In addition, we describe cutting-
edge analytic and informatic tools to study germ cell development at the single cell level and
meiotic recombination. The volume is divided into four sections. The first section is devoted
to methods for the isolation, purification, and transfection of PGCs, as well as methods for
the purification of GCs from the fetal human testes and the adult testes of mice. Section II is
v
11. divided into two parts.
systems for induction an
of the mouse germline
mammalian ovaries and
cells into pluripotent s
section focuses on descr
recombination initiation
bination events occurrin
The first part describes cytological methods for establishing in vitro
d culturing of PGCs from PGC-like cells and induction and editing
. The second part is dedicated to the isolation and culture of
oocytes. Section III describes a protocol for reprogramming somatic
tem cells (iPS) from patients with Klinefelter syndrome. The last
iptions of bioinformatic pipelines for studying GC development and
and a tool for image analysis of chromosome structure and recom-
g in prophase I of meiosis.
vi Preface
The editors thank the many authors of these chapters for their willingness to share their
protocols and expertise.
Rome, Italy Marco Barchi
Massimo De Felici
12. Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
PART I ISOLATION AND PURIFICATION OF PRIMORDIAL (AND) GERM CELLS
1 Isolation and Purification of Viable PGCs from Mouse Embryos. . . . . . . . . . . . . . 3
Massimo De Felici
2 Purification and Transfection Methods of Chicken Primordial Germ Cells . . . . . 15
Luiza Chojnacka-Puchta and Dorota Sawicka
3 Isolation and In Vitro Propagation of Human Spermatogonial
Stem Cells (SSCs). . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
Janmejay Hingu, Guillermo Galdon, Nicholas A. Deebel,
and Hooman Sadri-Ardekani
4 Purification by STA-PUT Technique of Male Germ Cells from Single
Mouse and RNA-Extraction for Transcriptomic Analysis. . . . . . . . . . . . . . . . . . . . . 37
Chiara Naro, Claudio Sette, and Raffaele Geremia
5 Isolation of Mouse Germ Cells by FACS Using Hoechst 33342
and SYTO16 Double Staining . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
Mark E. Gill, Hubertus Kohler, and Antoine H. F. M. Peters
6 Isolation and In Vitro Culture of Germ Cells and Sertoli Cells
from Human Fetal Testis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 63
Celine M. Roelse, Arend W. Overeem, Yolanda W. Chang,
Meriam Boubakri, and Susana M. Chuva de Sousa Lopes
PART II ESTABLISHING IN VITRO SYSTEMS FOR STUDYING PRIMORDIAL
(AND) GERM CELLS
7 Human Primordial Germ Cell-Like Cell Induction from Pluripotent
Stem Cells by SOX17 and PRDM1 Expression . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87
Naoko Irie, Toshihiro Kobayashi, and M. Azim Surani
8 Induction of Primordial Germ Cell-Like Cells from Rat Pluripotent
Stem Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99
Mami Oikawa, Masumi Hirabayashi, and Toshihiro Kobayashi
9 Induction of Meiotic Initiation in Long-Term Mouse Spermatogonial
Stem Cells Under Retinoid Acid and Nutrient Restriction Conditions. . . . . . . . . 113
Xiaoyu Zhang and Ning Wang
10 Lipofection-Based Delivery of CRISPR/Cas9 Ribonucleoprotein
for Gene Editing in Male Germline Stem Cells . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 123
Mariella Obermeier, Vera Rogiers, Tamara Vanhaecke, and Yoni Baert
11 Mouse In Vitro Spermatogenesis on 3D Bioprinted Scaffolds . . . . . . . . . . . . . . . . 135
Guillaume Richer, Tamara Vanhaecke, Vera Rogiers,
Ellen Goossens, and Yoni Baert
vii
13. viii Contents
12 Histological and Cytological Techniques to Study Perinatal Mouse
Ovaries and Oocytes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 151
Nikoleta Nikou, Maria L
opez Panadés, and Ignasi Roig
13 Method of Isolation and In Vitro Culture of Primordial Follicles
in Bovine Animal Model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 171
Pritha Dey, Noemi Monferini, Ludovica Donadini, Valentina Lodde,
Federica Franciosi, and Alberto Maria Luciano
PART III SOMATIC REPROGRAMMING TO STUDYING HUMAN ANEUPLOIDY
FEATURES
14 Generation of iPSC Cell Lines from Patients with Sex Chromosome
Aneuploidies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 185
Veronica Astro and Antonio Adamo
PART IV INFORMATIC TOOLS FOR STUDYING GERM CELLS DEVELOPMENT
AND RECOMBINATION
15 Data Analysis Pipeline for scRNA-seq Experiments to Study Early
Oogenesis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 203
Wei Ge, Teng Zhang, Yang Zhou, and Wei Shen
16 Mapping Meiotic DNA Breaks: Two Fully-Automated Pipelines
to Analyze Single-Strand DNA Sequencing Data, hotSSDS
and hotSSDS-extra. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 227
Pauline Auffret, Bernard de Massy, and Julie A. J. Clément
17 “MeiQuant”: An Integrated Tool for Analyzing Meiotic Prophase I
Spread Images. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 263
Julien Cau, Laurine Dal Toe, Akbar Zainu, Frédéric Baudat,
and Thomas Robert
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287
14. Contributors
ANTONIO ADAMO • Biological and Environmental Science and Engineering Division, King
Abdullah University of Science and Technology, Thuwal, Saudi Arabia
VERONICA ASTRO • Biological and Environmental Science and Engineering Division, King
Abdullah University of Science and Technology, Thuwal, Saudi Arabia
PAULINE AUFFRET • Ifremer, IRSI, SeBiMER Service de Bioinformatique de l’Ifremer,
Plouzané, France
M. AZIM SURANI • Wellcome Trust/Cancer Research U.K. Gurdon Institute, Henry Wellcome
Building of Cancer and Developmental Biology, Cambridge, UK; Physiology, Development
and Neuroscience Department, University of Cambridge, Cambridge, UK
YONI BAERT • Biology of the Testis Lab (BITE) Research Group, Department of Reproduction,
Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB), Brussels, Belgium;
In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit Brussel (VUB),
Brussels, Belgium
FRÉDÉRIC BAUDAT • Institut de Génétique Humaine, University of Montpellier, CNRS,
Montpellier, France
MERIAM BOUBAKRI • Department of Anatomy and Embryology, Leiden University Medical
Center, Leiden, The Netherlands
JULIEN CAU • Biocampus Montpellier, University of Montpellier, CNRS, INSERM,
Montpellier, France
YOLANDA W. CHANG • Department of Anatomy and Embryology, Leiden University Medical
Center, Leiden, The Netherlands
LUIZA CHOJNACKA-PUCHTA • Department of Chemical, Biological and Aerosol Hazards,
Central Institute for Labour Protection-National Research Institute, Warsaw, Poland
SUSANA M. CHUVA DE SOUSA LOPES • Department of Anatomy and Embryology, Leiden
University Medical Center, Leiden, The Netherlands; Department for Reproductive
Medicine, Ghent University Hospital, Ghent, Belgium
JULIE A. J. CLÉMENT • Institut de Génétique Humaine (IGH), Centre National de la
Recherche Scientifique, UnivMontpellier, Montpellier, France; IHPE, Univ Montpellier,
CNRS, IFREMER, Univ Perpignan Via Domitia, Perpignan, France
MASSIMO DE FELICI • Department of Biomedicine and Prevention, Section of Histology and
Embryology, Faculty of Medicine and Surgery, “Tor Vergata” University of Rome, Rome,
Italy
BERNARD DE MASSY • Institut de Génétique Humaine (IGH), Centre National de la
Recherche Scientifique, UnivMontpellier, Montpellier, France
NICHOLAS A. DEEBEL • Department of Urology, Wake Forest University School of Medicine,
Winston-Salem, NC, USA; Wake Forest Institute for Regenerative Medicine, Wake Forest
University School of Medicine, Winston-Salem, NC, USA
PRITHA DEY • Reproductive and Developmental Biology Laboratory, Department of
Veterinary Medicine and Animal Sciences, University of Milan, Milan, Italy
LUDOVICA DONADINI • Reproductive and Developmental Biology Laboratory, Department of
Veterinary Medicine and Animal Sciences, University of Milan, Milan, Italy
FEDERICA FRANCIOSI • Reproductive and Developmental Biology Laboratory, Department of
Veterinary Medicine and Animal Sciences, University of Milan, Milan, Italy
ix
15. x Contributors
GUILLERMO GALDON • Wake Forest Institute for Regenerative Medicine, Wake Forest
University School of Medicine, Winston-Salem, NC, USA
WEI GE • College of Life Sciences, Qingdao Agricultural University, Qingdao, China
RAFFAELE GEREMIA • Department of Biomedicine and Prevention, Section of Human
Anatomy, University of Rome Tor Vergata, Rome, Italy
MARK E. GILL • Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland;
Reproductive Medicine and Gynecological Endocrinology, University Hospital Basel, Basel,
Switzerland
ELLEN GOOSSENS • Biology of the Testis Lab (BITE) Research Group, Department of
Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB),
Brussels, Belgium
JANMEJAY HINGU • Department of Urology, Wake Forest University School of Medicine,
Winston-Salem, NC, USA; Wake Forest Institute for Regenerative Medicine, Wake Forest
University School of Medicine, Winston-Salem, NC, USA
MASUMI HIRABAYASHI • Center for Genetic Analysis of Behavior, National Institute for
Physiological Sciences, Aichi, Japan; The Graduate University of Advanced Studies, Aichi,
Japan
NAOKO IRIE • Wellcome Trust/Cancer Research U.K. Gurdon Institute, Henry Wellcome
Building of Cancer and Developmental Biology, Cambridge, UK; Metabolic Systems
Laboratory, Live Imaging Center, Central Institute for Experimental Animals, Kawasaki-
ku, Kanagawa, Japan
TOSHIHIRO KOBAYASHI • Division of Mammalian Embryology, Center for Stem Cell Biology
and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo,
Minato-ku, Tokyo, Japan; Center for Genetic Analysis of Behavior, National Institute for
Physiological Sciences, Okazaki, Aichi, Japan
HUBERTUS KOHLER • Friedrich Miescher Institute for Biomedical Research, Basel,
Switzerland
VALENTINA LODDE • Reproductive and Developmental Biology Laboratory, Department of
Veterinary Medicine and Animal Sciences, University of Milan, Milan, Italy
MARIA LÓPEZ PANADÉS • Genome Integrity and Instability Group, Institut de Biotecnologia i
Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Cell
Biology, Physiology, and Immunology, Cytology and Histology Unit, Universitat Autònoma
de Barcelona, Barcelona, Spain
ALBERTO MARIA LUCIANO • Reproductive and Developmental Biology Laboratory,
Department of Veterinary Medicine and Animal Sciences, University of Milan, Milan,
Italy
NOEMI MONFERINI • Reproductive and Developmental Biology Laboratory, Department of
Veterinary Medicine and Animal Sciences, University of Milan, Milan, Italy
CHIARA NARO • Department of Neuroscience, Section of Human Anatomy, Catholic
University of the Sacred Heart, Rome, Italy; GSTeP-Organoids Research Core Facility,
Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
NIKOLETA NIKOU • Genome Integrity and Instability Group, Institut de Biotecnologia i
Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Cell
Biology, Physiology, and Immunology, Cytology and Histology Unit, Universitat Autònoma
de Barcelona, Barcelona, Spain
MARIELLA OBERMEIER • Biology of the Testis Lab (BITE) Research Group, Department of
Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB),
Brussels, Belgium
16. Contributors xi
MAMI OIKAWA • Division of Mammalian Embryology, Center for Stem Cell Biology and
Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo,
Japan; Laboratory of Regenerative Medicine, Tokyo University of Pharmacy and Life
Sciences, Tokyo, Japan
AREND W. OVEREEM • Department of Anatomy and Embryology, Leiden University Medical
Center, Leiden, The Netherlands
ANTOINE H. F. M. PETERS • Friedrich Miescher Institute for Biomedical Research, Basel,
Switzerland; Faculty of Science, University of Basel, Basel, Switzerland
GUILLAUME RICHER • Biology of the Testis Lab (BITE) Research Group, Department of
Reproduction, Genetics and Regenerative Medicine, Vrije Universiteit Brussel (VUB),
Brussels, Belgium
THOMAS ROBERT • CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS,
INSERM, Montpellier, France
CELINE M. ROELSE • Department of Anatomy and Embryology, Leiden University Medical
Center, Leiden, The Netherlands
VERA ROGIERS • In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije Universiteit
Brussel (VUB), Brussels, Belgium
IGNASI ROIG • Genome Integrity and Instability Group, Institut de Biotecnologia i
Biomedicina, Universitat Autònoma de Barcelona, Barcelona, Spain; Department of Cell
Biology, Physiology, and Immunology, Cytology and Histology Unit, Universitat Autònoma
de Barcelona, Barcelona, Spain
HOOMAN SADRI-ARDEKANI • Department of Urology, Wake Forest University School of
Medicine, Winston-Salem, NC, USA; Wake Forest Institute for Regenerative Medicine,
Wake Forest University School of Medicine, Winston-Salem, NC, USA
DOROTA SAWICKA • Department of Chemical, Biological and Aerosol Hazards, Central
Institute for Labour Protection-National Research Institute, Warsaw, Poland
CLAUDIO SETTE • Department of Neuroscience, Section of Human Anatomy, Catholic
University of the Sacred Heart, Rome, Italy; GSTeP-Organoids Research Core Facility,
Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
WEI SHEN • College of Life Sciences, Qingdao Agricultural University, Qingdao, China
LAURINE DAL TOE • CBS (Centre de Biologie Structurale), Univ Montpellier, CNRS,
INSERM, Montpellier, France
TAMARA VANHAECKE • In Vitro Toxicology and Dermato-Cosmetology (IVTD), Vrije
Universiteit Brussel (VUB), Brussels, Belgium
NING WANG • Department of Cell Biology and Physiology, University of Kansas Medical
Center, Kansas City, KS, USA; Center for Reproductive Sciences, Institute for
Reproductive and Developmental Sciences (IRDS), University of Kansas Medical Center,
Kansas City, KS, USA
AKBAR ZAINU • Institut de Génétique Humaine, University of Montpellier, CNRS,
Montpellier, France
TENG ZHANG • College of Life Sciences, Inner Mongolia University, Hohhot, China
XIAOYU ZHANG • Department of Cell Biology and Physiology, University of Kansas Medical
Center, Kansas City, KS, USA; Center for Reproductive Sciences, Institute for
Reproductive and Developmental Sciences (IRDS), University of Kansas Medical Center,
Kansas City, KS, USA
YANG ZHOU • College of Life Sciences, Inner Mongolia University, Hohhot, China
19. niche. From about 13.5 dpc, prospermatogonia undergo a tempo-
rary GO mitotic arrest while oogonia enter the prophase of meiosis
I as primary oocytes.
4 Massimo De Felici
PGCs representing a key cell type for analysis of germ-soma
differentiation as well as pluripotency at the molecular level. More-
over, in the last years, the possibility to generate primordial germ
cells–like cells (PGCLCs) in vitro able to generate male and female
gametes from pluripotent stem cells of various mammalian species
including humans (for a review, see Ref. [3]) increased exponen-
tially the interest about such unique cell type. Today single-cell
genome and transcriptome sequencing methods enabling the
simultaneous molecular analysis of hundreds, or thousands of
cells, inside heterogeneous populations are considerably increasing
our capability to analyze the PGC genome and transcriptome (for
example, see Refs. [4–6]. Nevertheless, for many laboratories, the
isolation and purification methods of PGCs at different stages from
embryos remain the most shortcut and affordable tool to study
many aspects of their development at cellular and molecular levels.
Historically, the isolation and purification of PGCs from mouse
embryos between 8.5 and 12.5 dpc were attempted using cellular,
molecular, and transgenic approaches in parallel with the progress
of technologies as summarized in Table 1. At these stages, PGCs are
characterized by motility capability and active proliferation; they are
for the most part single or form small clusters and do not form firm
contacts with the surrounding extracellular matrix molecules and
surrounding somatic cells. Both female and male mouse PGCs
express surface markers such as tissue-nonspecific alkaline phospha-
tase (TNAP), the carbohydrate epitopes bearing terminal Lewis X
determinants (Stage-specific embryonic antigen-1 (SSEA-1) and
the antigenic determinant of monoclonal antibody EMA-1, E-cad-
herin, and the c-Kit and CXCR4 receptors. In principle, antibodies
against all these surface molecules can be used to purify PGCs at
these stages. Moreover, PGCs express several genes typical of plu-
ripotent stem cells such as Pou5f1 (Oct4), Sox2, Nanog and of the
germline such Nanos3, Dazl and Ddx4 (Vasa). Therefore, PGCs
can be purified also from embryos of transgenic mice carrying
reporter genes under the control of such genes (for example, see
Refs. [12, 13]).
In the present chapter, I focus on immunomagnetic cell sorting
(iMACS) and fluorescence-activated cell sorting (FACS) methods
used in my laboratory for the purification of mouse PGCs from
10.5 to 12.5 dpc embryos using anti-SSEA-1 (CD15) MicroBeads
and the transgenic mouse line Oct4ΔPE: eGFP (OG2),
respectively.
20. Purification of Mouse PGCs 5
Table 1
Timetable of the methods used for the first time to isolate and purify mouse PGCs
Heath J. K. (1978) [7] (Cell 15, 299–306) Manual dissection of gonadal ridges
De Felici M. and McLaren A. (1982) [8] Percoll gradient
McCarrey J. R. et al. (1987) [9] Immunopurification by FACS
Massimo De Felici and Maurizio Pesce (1995) [10] Immunoaffinity adhesion
Pesce, M., and De Felici, M. (1995) [11] Immunopurification by MACS
Abe K. et al. (1996) [12] (Dev. Biol. 189, 468–472) Transgenic TNAPβ-geo
PGCs by FACS-gal
Szabò P. E. et al. (2002) [13] Transgenic Oct4ΔPE:eGFP PGCs by FACS
Mayanagi T. et al. (2003) [14] Nicodenz gradient
2 Materials and Instruments
2.1 Isolation of PGCs
by MiniMACS
1. 10.5–12.5 dpc pregnant females of desired genetic background
(see Note 1).
2. 1xPBS, sterile, for dissecting.
3. Hanks’ Balanced Salt Solution (HBSS) or M2 (Sigma-Aldrich).
4. Trypsin/EDTA solution (0.025% trypsin and 0.01% EDTA) in
PBS (Gibco).
5. DNase (1 mg/mL) solution (STEMCELL Technology).
6. Culture-tested fetal calf serum (FCS).
7. Poly-L-lysine-coated glass coverslips: soak coverslips in
200 μg/mL poly-L-lysine in distilled water for at least 1 h at
r. t., wash three times in distilled water and air-dry.
8. Chromogenic solution for alkaline phosphatase
(AP) detection: immediately before use dissolve 1 mg/mL
fast red TR salt or fast blue BB salt (Sigma-Aldrich) in distilled
water and add 40 μL/mL naphthol AS-BI or AS-MX alkaline
solution (Sigma-Aldrich).
9. Fine scissors, dissection forceps and No. 5 watchmaker’s for-
ceps, sterile.
10. Eppendorf or equivalent single channel 10–100 μL pipette.
11. 18- and 23-gauge needle and 1 mL syringe for tissues
dissection.
12. 5 mL test tubes and 1.5 and 2 mL Eppendorf tubes.
13. Stereomicroscope (the author uses a Wild M5a from Leica,
maximum magnification 100×, equipped with both incident
and transmitted light sources, the latter preferably with a mov-
able mirror).
21. 6 Massimo De Felici
14. Anti-SSEA-1 (CD15) MicroBeads (130-094-530, Miltenyi
Biotech).
15. MACS buffer: phosphate-buffered saline (DPBS) without
Ca2
+ and Mg2
+, with 0.5% bovine serum albumin (BSA), and
2 mM EDTA (keep buffer cold at 2-8 °C and degas before
use, as air bubbles could block the column).
16. MiniMACS™ Separator (Miltenyi Biotech).
17. MACS MultiStand (Miltenyi Biotech).
18. MACS MS separation columns (Miltenyi Biotech).
19. Class II Biological Hazard laminar flow hood (see Note 2).
2.2 Isolation of PGCs
from OG2 Mice by
FACS
Materials listed in the previous section from 2 to 11 are required
also in this procedure.
1. 10.5–12.5 dpc pregnant CD1 females previously mated with
OG2 homozygous males (see Note 3).
2. Hanks’ Balanced Salt Solution (HBSS) or M2 (Sigma-Aldrich)
plus 1 mg/mL BSA.
3. 5 mL FACS tubes compatible with flow cytometer.
4. Sorting flow cytometer (see Note 4).
3 Methods
3.1 Dissection of
AGM and GRs
At 10.5 dpc, the region dissected away from the embryo (hindgut,
dorsal mesentery, and gonadal ridges) corresponds to the region
termed AGM (aorta, gonad, mesonephros) containing the most
part of migratory PGCs (about 1000/embryo, while at 11.5 and
12.5 dpc PGCs are obtained from the gonadal ridges (GRs, about
3000/and 8000/embryo) ([15], and our unpublished data).
Embryonic sex can be determined only by PCR of genomic DNA
using specific primers for 10.5 and 11.5 dpc samples (see Note 5) or
by assessing gonad morphology for 12.5 dpc (Fig. 1).
The procedure described in the following steps is routinely
used in the author’s laboratory, but it can be varied according to
the operator’s preference and experience.
Steps
1. Lay the freshly killed animal on its back and soak the abdomen
with 70% alcohol. CD1 mice, usually employed in the author’s
laboratory, were housed and mated under conventional condi-
tions (see Note 1). Positive plug test is considered 0.5-day post
coitum. The developmental stage of embryos can be precisely
22. Purification of Mouse PGCs 7
Fig. 1 (a) Morphology of limb buds of the mouse embryo between 10.5 and 12.5 dpc. (b) Sex indifferent
11.5 dpc gonadal ridges (upper), 12.5 dpc testis (right), 12.5 dpc ovary (left)
determined by observing the morphology of the hind limb bud
(Fig. 1).
2. Pinch up the skin of the abdomen region between thumb and
forefinger of both hands and pull it toward the head and tail
until the abdomen is completely exposed and the fur is well out
of the way.
3. Using forceps and fine scissors, cut the body wall, push the coil
of gut out of the way, and locate the two horns of the uterus.
4. Remove the uterus intact by cutting across the cervix and the
two uterotubal junctions and trim away the fat and mesentery
with fine scissors.
5. Transfer the uterus in a petri dish in a volume of PBS sufficient
to completely immerse the tissues and remove any residual fat
and mesentery.
6. Transfer the uterus to a second dish of fresh medium and
dissect the embryos with attached placenta by cutting the anti-
mesometrial wall of the uterus with the tips of scissors. Remove
the embryo’s membranes by cutting at the junction of the
placenta with watchmaker’s forceps.
7. Cut with scissors the anterior half of the embryo just below the
armpits and transfer the posterior half to a dish of fresh
medium,
8. Under a stereomicroscope, turn back such posterior half and
hold it with No. 5 watchmaker’s forceps while making a cut
along the ventral midline.
9. For 10.5 dpc embryos, using the tips of No. 5 watchmaker’s
forceps, remove viscera paying attention does not remove the
23. 8 Massimo De Felici
dorsal mesentery and the hindgut. Peel away these and asso-
ciated GRs (gonad + mesonephros) from the dorsal region of
the embryo and transfer them into HBSS or M2 (Fig. 2).
10. For 11.5 and 12.5 dpc embryos, scoop out liver and any
remnants of intestine with No. 5 watchmaker’s forceps and
identify the GRs lying on the dorsal wall of the embryos on
either side of the abdominal aorta. Slide the tips of the
No. 5 watchmaker’s forceps behind each GR and remove
them from the embryo.
11. Transfer the GRs to HBSS or M2 and separate the gonads from
mesonephroi using 25G needles.
3.2 Isolation of 10.5–
12.5 dpc PGCs by
MiniMACS
In this method, PGCs are separated from the other cell types by
positive selection. This means that they are labeled with superpar-
amagnetic microbeads conjugated to highly specific antibodies.
During separation, the magnetically labeled PGCs are retained
within a column, while unlabeled cells flow through. After a wash-
ing step, the column is removed from the magnetic field of the
separator, and PGCs are eluted. Positive selection can be performed
by direct or indirect magnetic labeling. The following procedure
describes a direct labelling of PGCs by anti-SSEA-1 MicroBeads
purchased from Miltenyi Biotechnology.
The simplicity and low cost make the MiniMACS separation
procedure the method of choice for rapidly obtaining reasonable
numbers of purified germ cells and somatic cells that can be used
immediately for biochemical studies or be grown in suitable in vitro
culture systems. In 2–3 h of work, one person should be able to
isolate from 10 embryos of 10.5, 11.5, and 12.5 dpc (a reasonable
yield from a pregnant CD1 female) about 5000–7000 (purity
70–80%), 10,000–15,000, and 40,000–50,000 PGCs (purity
85–95%), respectively (Fig. 3). The presence of supermagnetic
microbeads coated with antibodies on the cell surface might be
harmful for certain cell functions. However, we cultured PGC
purified by MiniMACS for several days without any apparent dele-
terious effect (for example see Ref. [16]).
Steps
1. 10.5 dpc AGM (usually 10–20) or 11.5–12.5 gonads (usually
20–40) are transferred with 100 μL Eppendorf pipette in mini-
mal amount of medium into a 1.5 mL Eppendorf tube contain-
ing 0.5 mL of trypsin-EDTA.
2. After about 5 min incubation at 37 °C carefully remove the
solution and wash the tissues twice with 1 mL of HBSS or
M2 + 5% FCS, allowing them to settle on the bottom of the
tube. Leave the tissue in about 100 μL of medium and pipette
up and down with the Eppendorf pipette for their complete
24. Purification of Mouse PGCs 9
Fig. 2 Dissected AGM (aorta, gonad, mesonephros) from 10.5 dpc embryo
Fig. 3 Mouse 12.5 dpc PGCs stained with anti-SSEA-1 (CD15) FITC antibodies
dispersion (about 10 strokes are usually sufficient). Ensure a
single cell suspension by passing through an 18- and 23-gauge
needle.
3. Add 1 mL of MACS buffer containing 10 μg/mL DNase,
centrifuge at 500 ×g for 10 min at 4 °C. Remove supernatant
and resuspend the pellet in 80 μL of buffer with the Eppendorf
pipette (see Note 6).
4. Add 20 μL anti-SSEA-1 MicroBeads and incubate with contin-
uous agitation for 45–60 min at 4 °C.
5. Dilute the cells to a final volume of 1.5 mL with MACS buffer
and transfer on top of an AS column previously washed, filled
with the buffer, and placed in the magnetic field of a Mini-
MACS separation unit (Fig. 4).
6. Collect the effluent as SSEA-1 negative cells (somatic cells) and
wash the column twice with 1 mL buffer into 5 mL
collecting tube.
7. Remove the column from the magnetic field and, using the
plugger supplied with the column, flush out with 1.5 mL buffer
the cells retained by the column (SSEA-1 positive PGCs) in a
2 mL Eppendorf tube.
25. 10 Massimo De Felici
Fig. 4 A MiniMACS separation unit form Miltenyi Biotec
8. To assess PGC purity, a small sample of the cell suspension
(20–30 μL) can be transferred onto a polylysinated coverglass
and stained for AP.
3.3 Isolation of 10.5–
12.5 dpc PGCs by FACS
At these developmental stages, FACS can be used to purify viable
PGCs labeled with antibodies anti-SSEA-1 (CD15) or other sur-
face markers (e.g., EMA-1, E-cadherin, Kit) conjugated with vari-
ous fluorochromes commercially available from several companies.
Alternatively, PGCs can be isolated from transgenic mice expressing
reporter genes under the control of early PGCs genes (e.g., Pou5f1,
Prdm1, and Dppa3). Here we describe PGC purification from OG2
transgenic mice (Oct4ΔPE:eGFP) expressing enhanced green fluo-
rescent protein (eGFP) under the control of the Pouf1 (Oct4),
promoter and distal enhancer [17] (Fig. 5a).
Steps
1. After steps 1 and 2 of Subheading 3.2 Isolation of PGCs by
MiniMACS, add 1 mL of HBSS plus 1 mg/mL BSA contain-
ing 10 μg/mL DNase, centrifuge at 500 ×g for 10 min at 4 °C.
Remove supernatant, and resuspend the pellet in 0.5 mL of
HBSS with the Eppendorf pipette (see Note 6).
2. Transfer the cell suspension into a FACS tube on ice and
proceed to flow cytometer.
3. Cell debris and dead cells are excluded from the analysis based
on scatter signals and propidium iodide fluorescence. Viable
cells are gated based on forward scatter (FSC) vs. side scatter
(SSC) profile. eGFP vs. FSC will delineate eGFP-positive PGC
26. Purification of Mouse PGCs 11
Fig. 5 (a) 12.5 dpc ovary and testis from OG2 mouse. Note a spotty pattern of fluorescence in the ovary,
whereas a wavy pattern reflecting testicular cords formation is visible in the testis. (b) Representative FACS
profile of disaggregated gonads isolated from 12.5 dpc CD1/OG2 embryos. Right: FSC-A/SSC-A dot plots of
the cell suspension showing the cell population selected for fluorescence analysis. Left: GFP-A/FSC-A showing
two eGFP positive PGC populations to be sorted displaying high or dull fluorescence; Inset: a group of PGCs
positive for eGFP-OCT4
population of cells and the remaining eGFP negative cells as
somatic cells (Fig. 5b), (see Note 7).
4. To assess PGC purity, a small sample of the cell suspension
(20–30 μL) can be transferred onto a polylysinated coverglass
and observed onto a fluorescence phase contrast microscope.
The purity of sorted PGC population was usually 98%.
5. Sorted cells can be used immediately for biochemical studies or
be grown in suitable in vitro culture systems.
27. )
12 Massimo De Felici
4 Notes
1. Use the mouse strain of your choice, although we have fond
that outbred strains such as MF1 and CD1 mice give larger
numbers of PGCs than some inbred strains (e.g., BalbC). The
procedure described here and the mean number of PGCs
obtained are from CD1 embryos. Moreover, consider that
PGC number can vary considerably among embryos.
2. The safety cabinet is necessary to carry out the whole procedure
if sterility must be maintained.
3. The OG2 mice can be purchased from Jackson Laboratory, Cat
# 003715. To ensure large litter sizes and large numbers of
germ cells per gonad homozygous OG2 male can be mate with
CD1 females. For the present protocol, usually, a minimum of
two litters is pooled.
4. For the present procedure, cell sorting was performed on a
FACSAria II (Becton Dickinson) interfaced with the FACS-
DiVa 6.0 Software.
5. If sexing is required, each embryo is dissected into a 24-well
plate containing warm DMEM+10% FCS and a tail tip is taken
for PCR sexing. We used genomic PCR of mouse Ube1 [18] or
Xlr and Sly [19] genes. Once results are available (2–3 h
embryos are pooled based on sex.
6. For optimal performance, it is important to obtain a single-cell
suspension. Check a small sample of the cell suspension under a
microscope and if necessary, pass the suspension through a
23-gauge needle once more or filter through a 40 μm cell
strainer (BD Falcon). The percent of PGCs in the cell suspen-
sion estimated by APase staining (see step 8) was less than 0.1%
and approximately 5% and 30–35% at 10.5, 11.5, and 12.5 dpc,
respectively.
7. Although the most part of PGCs showed high eGFP, we and
others observed a little population of weakly eGFP positive
cells. At 10.5 and 11.5 dpc, these latter represented between
9% and 12% of the whole eGFP positive cells. Hypotheses
about such minority PGC population are discussed in
[20]. Although FACS is the method of choice for cell sorting
when high cell purity is needed, it suffers, however, from
(sometimes very) high losses, requires trained personnel, and
is expensive to own and operate. The estimation of PGC recov-
ery under the present conditions varied from 65% to 70%. This
means that from 10 embryos of 10.5, 11.5, and 12.5 dpc
heterozygotes CD1/OG2 females, about 5000, 15,000, and
35,000 PGCs can be obtained, respectively. Finally, PGCs
28. Purification of Mouse PGCs 13
isolated by FACS from transgenic fluorescent proteins should
not have any potential adverse effect caused by antibodies
bound to their cell surface.
References
1. De Felici M (2016) The formation and migra-
tion of primordial germ cells in mouse and
man. Results Probl Cell Differ 58:23–46
2. Mikedis MM, Downs KM (2014) Mouse pri-
mordial germ cells: a reappraisal. Int Rev Cell
Mol Biol 14(309):1–57
3. Hayashi M, Kawaguchi T, Durcova-Hills G,
Imai H (2017) Generation of germ cells from
pluripotent stem cells in mammals. Reprod
Med Biol 17:107–114
4. Guo F, Yan L, Guo H, Lin Li L et al (2015)
The transcriptome and DNA methylome land-
scapes of human primordial germ cell. Cell
161:1437–1452
5. Zhao ZH, Ma JY, Meng TG, Wang ZB et al
(2020) Single-cell RNA sequencing reveals the
landscape of early female germ cell develop-
ment. FASEB J 34:12634–12645
6. Ge W, Wang JJ, Zhang RQ, Tan SJ et al (2021)
Dissecting the initiation of female meiosis in
the mouse at single-cell resolution. Cell Mol
Life Sci 78:695–713
7. Heath JK (1978) Characterization of a xeno-
geneic antiserum raised against the fetal germ
cells of the mouse: cross-reactivity with embry-
onal carcinoma cells. Cell 15:299–306
8. De Felici M, McLaren A (1982) Isolation of
mouse primordial germ cells. Exp Cell Res
142:476–482
9. McCarrey JR, Hsu KC, Eddy EM, Klevecz RR,
Bolen JL (1987) Isolation of viable mouse pri-
mordial germ cells by antibody-directed flow
sorting. J Exp Zool 242:107–111
10. De Felici M, Pesce M (1995) Immunoaffinity
purification of migratory mouse primordial
germ cells. Exp Cell Res 26:277–279
11. Pesce M, De Felici M (1995) Purification of
mouse primordial germ cells by MiniMACS
magnetic separation system. Dev Biol 170:
722–725
12. Abe K, Hashiyama M, Macgregor G, Yama-
mura K (1996) Purification of primordial
germ cells from TNAPbeta-geo mouse
embryos using FACS-gal. Dev Biol 180:468–
472
13. Szabò PE, Hübner K, Schöler H, Mann JR
(2002) Allele-specific expression of imprinted
genes in mouse migratory primordial germ
cells. Mech Dev 115:157–160
14. Mayanagi T, Kurosawa R, Ohnuma K, Ueyama
et al (2003) Purification of mouse primordial
germ cells by Nycodenz. Reproduction 125:
667–675
15. Tam P, Snow M (1981) Proliferation and
migration of primordial germ cells during com-
pensatory growth in mouse embryos. J Morph
Exp Morph 64:133–147
16. Farini D, Scaldaferri ML, Iona S, La Sala G, De
Felici M (2005) Growth factors sustain primor-
dial germ cell survival, proliferation and enter-
ing into meiosis in the absence of somatic cells.
Dev Biol 285:49–56
17. Yoshimizu T, Sugiyama N, De Felice M, Yeom
YI et al (1999) Germline-specific expression of
the Oct-4/green fluorescent protein (GFP)
transgene in mice. Dev Growth Differ 41:
675–684
18. Chuma S, Nakatsuji N (2001) Autonomous
transition into meiosis of mouse fetal germ
cells in vitro and its inhibition by gp130-
mediated signaling. Dev Biol 229:468–479
19. McFarlane L, Truong V, Palmer JS, Wilhelm D
(2013) Novel PCR assay for determining the
genetic sex of mice. Sex Dev 7:207–211
20. Scaldaferri ML, Klinger FG, Farini D, Di Carlo
A et al (2015) Hematopoietic activity in puta-
tive mouse primordial germ cell populations.
Mech Dev 136:53–63
30. [5]. Considering such dynamics localization, at the 10 HH stage,
PGCs can be obtained from blastoderm, at 13–17 HH stages (2.5-
to 3-day-old embryos) from blood and at stages 26–29 HH stages
(5.5- to 6-day-old embryos) directly from the embryonic gonads.
PGCs isolated from these stages have been successfully used for
transfection [4, 5] or cryoconservation [6].
16 Luiza Chojnacka-Puchta and Dorota Sawicka
Purification methods for chick PGCs range from density gra-
dients using Nycodenz [7], Ficoll [8], or Percoll [9] to immuno-
magnetic cell sorting (MACS) and fluorescence-activated cell
sorting (FACS) [11]. Chick PGCs were purified from blood using
ammonium chloride-potassium (ACK) lysis buffer to eliminate red
blood cells, followed by in vitro culture [10]. Using such methods,
the purity of PGCs overcame 90% at best.
Chick PGCs can be identified based on morphological criteria,
i.e., large size and presence of a large amount of glycogen in the
cytoplasm [12]. Glycogen granules can be evidenced also by peri-
odic acid-Schiff (PAS) staining [13, 14]. Moreover, chick PGCs
were stained for alkaline phosphatase activity, a classical PGC
marker also in mammalian species [15], and for many immunolog-
ical epitopes for surface glycoproteins. The best known and char-
acterized is SSEA-1 (Stage-specific Embryonic Antigen-1) [16],
which is effective only after stage 10 HH. Other immunological
markers are EMA-1 (Epithelial Membrane Antigen) [17] expressed
from the crescent stage [18] and MVH (Mouse Vasa Homolog)
[19]. Interestingly, all these immunological markers are common
to mammalian PGCs (see Chap. 1, by M. De Felici, in the
present book)
Here we describe a simple and rapid method based on Percoll
density gradient suitable to purify chicken PGC from blood and
early embryonic gonads that can be successfully transfected using
the electroporation/lipofection method for germline manipula-
tion. Details are in Table 1.
Table 1
Average numbers of chicken embryos, PGCs isolated from blood and gonads, purity and efficiency of
transfection methods
Type
of
PGCs
Number
of
embryosa
Number of total
cells after
isolation
Number of total
after Percoll
purification
Purity of
PGCs
(%)
Electroporation
efficiency (%)
Lipofection
efficiency
(%)
bPGCs 60 11.6 × 106
1.76 × 106
64 75 50
gPGCs 30 4.53 × 106
7.56 × 105
N/A 73 39
Data obtained in our experiments
a
The number of embryos used to isolate PGCs depends on the quality of embryos and operator skills. Some of them can
be unfertilized or undeveloped
31. Purification and Transfection of PGCs 17
2 Materials
2.1 Reagents 1. Freshly fertilized eggs.
2. 0.2 M EDTA: Prepare 100 mL of 0.2 M EDTA by solving
7.44 g EDTA in 100 mL distillated water. Mix and adjust
pH 7.4 with NaOH. This solution must be autoclaved. Prepare
10 mL of 0.1 mM EDTA by dilution of 5 μL 0.2 M EDTA in
10 mL 1× PBS (without calcium or magnesium) and mix.
3. 0.25% trypsin EDTA: Dilute 40 μL 0.25% trypsin EDTA solu-
tion in 960 μL OptiMEM I to obtain 0.01% trypsin; make fresh
every time.
4. Antibiotic: Penicillin/streptomycin (10,000 U/mL).
5. OptiMEM I supplemented with 5% FBS. For 50 mL: add
2.5 mL FBS to 47.5 mL of OptiMEM I. Store at 4 °C.
6. Percoll 100% to prepare density solutions: 50%, 25%, and
12.5%. Make the steps by diluting the 100% Percoll, as shown
in Table 2. All solutions are freshly made; phenol red is used to
provide visual contrast for different phases.
7. Culture complex medium: OptiMEM I, 2% CS—chicken
serum, 10% FBS—fetal bovine, 20 ng/mL bFGF—basic fibro-
blast growth factor, 9 ng/mL mLIF—murine leukemia inhibi-
tory factor, 5 ng/mL hSCF—human stem cell factor, 1%
antibiotic (and selective antibiotic).
2.2 Equipment 1. Egg incubator.
2. Cell tram air/oil pump with capillary holder and fine glass
micropipette (90 × 135 mM).
3. Dissection microscope (a binocular stereoscopic dissection
microscope is preferred).
4. Centrifugator with swinging-bucket rotor and buckets.
5. CO2 incubator (5%).
6. Cell counter/hemocytometer.
Table 2
Steps to prepare Percoll solutions
Step Percoll final concentration (%) OptiMEM I with 1% FBS (mL) 100% Percoll (mL)
1. 50 10 10
2. 25 15 5
3. 12.5 17.5 2.5
The amounts of medium OptiMEM I with 5% FBS and Percoll are calculated for a final volume of 20 mL
32. 18 Luiza Chojnacka-Puchta and Dorota Sawicka
7. 15 mL tubes, 1.5 mL Eppendorf tubes.
8. Inverted/fluorescent microscope.
9. 4-well plates, petri dishes.
10. Electroporator, 0.4 mL cuvettes.
3 Methods
Prepare a workplace, where cells will be isolated from chicken
embryos by wiping the table with a disinfectant or 70% ethanol
solution. Also wipe the elements of the stereoscope and cell tram
air/oil pump. Perform all procedures using gloves and wear a
lab coat.
3.1 Preparation
Embryos to Isolate
of PGCs
1. Incubation eggs.
Incubate freshly fertilized laid chicken eggs (egg or meat breed)
at 37.8 °C and 60–62% relative humidity for 50–56 h to achieve
chicken embryos at 14–16 HH and for 6 days to obtain chicken
embryos at 28–29 HH (see Note 1).
3.2 Isolation of PGCs
from Bloodstream
(bPGCs)
1. Prepare 0.5 mL of 0.1 mM EDTA solution in 1 × PBS and
supplement with 1% antibiotic. Prepare the cell tram air/oil
pump and put the fine glass micropipette into a capillary
holder. Fill the tip of the fine glass micropipette with the
prepared solution. EDTA solution will prevent the drawn
blood from clotting.
2. Take out chicken eggs from the incubator and wipe with a 70%
solution of ethanol (see Note 2).
3. Gently transfer the contents of the egg to a sterile petri dish. Be
careful not to damage the embryo. Transfer the petri dish
under the stereoscope.
4. Take blood from the dorsal aorta of embryos (HH stages
14–16) using fine glass micropipette and suspend into EDTA
solution to obtain suspension-contained bPGCs and blood
counts (Fig. 1). Afterward, centrifuge cell suspension
(400 × g, 5 min) and resuspend in medium (0.5 mL) Opti-
MEM I supplemented with 5% FBS and 1% antibiotic.
5. Stain cells with 0.4% trypan blue (1:1) and count in a hemocy-
tometer under an inverted microscope.
3.3 Isolation of PGCs
from Gonads (gPGCs)
1. Prepare 1 × PBS containing 1% antibiotic at room
temperature (RT).
2. Take chicken eggs from incubator and wipe with a 70% ethanol
solution.
33. Purification and Transfection of PGCs 19
Fig. 1 Suspension of cells isolated from bloodstream of chicken embryos (HH stages 14–16) containing bPGCs
and blood counts (20×)
Fig. 2 Steps/scheme of isolation suspension of cells including gPGCs from gonads of 6-day-old chicken
embryos. (a) chicken embryo on the 6-th day of incubation, (b) mesonephros with gonads (6.3×), (c) isolated
gonads (40×), (d) suspension of cells containing gPGCs (20×)
3. Transfer the embryo (28–29 HH) (Fig. 2a) to a petri dish and
purify the remains of the yolk by washing several times with
1 × PBS containing 1% antibiotic.
4. Transfer the embryo to a clean petri dish and isolate mesoneph-
ros under a stereoscope (Fig. 2b).
5. Cut along the inner edge of the mesonephros with a 0.4 mM
blade to isolate gonads (Fig. 2c) and transfer gonads into the
tube with 1 × PBS containing 1% antibiotic.
34. 20 Luiza Chojnacka-Puchta and Dorota Sawicka
6. Mash the gonads with an insulin needle to increase the surface
available for the enzyme.
7. Pipette vigorously and transfer all content to the new tube.
8. Centrifuge (400 × g, 5 min).
9. Add 1 mL of 0.01% trypsin and digest for 1 min at 37 °C in a
heating block placed in a laminar airflow chamber. Inactivate
the enzyme by adding 100 μL of FBS.
10. Centrifuge suspension of cells, including gPGCs (400 × g,
5 min), and resuspend in 0.5 mL medium OptiMEM I supple-
mented with 5% FBS and 1% antibiotic. The obtained cells are
presented in Fig. 2d.
11. Stain cells with 0.4% trypan blue (1:1) and count in a hemocy-
tometer under an inverted microscope.
3.4 Purification and
Identification of PGCs
1. Prepare Percoll solutions: 50%, 25%, and 12.5% in medium
OptiMEM I supplemented with 5% FBS. Form density gradi-
ent by layering 3 mL each of 50%, 25%, and 12.5% density of
Percoll in a clear 15 mL conical centrifuge tube (see Note 3).
3.4.1 Percoll Purification
2. Suspend cells isolated from the bloodstream or gonads in 3 mL
of OptiMEM I with 5% FBS.
3. Place suspension of cells (3 mL) on the top of the density
gradient (Fig. 3) and centrifuge at 1.160 g for 20 min at 20 °
C (see Note 4).
4. Gently collect bPGCs or gPGCs between 50% and 25% Percoll
layers (see Note 5).
5. Wash PGCs twice in medium OptiMEM I (with 5% FBS, 1%
antibiotic).
6. Resuspend the cell pellet in medium OptiMEM I (with 5%
FBS, 1% antibiotic), and centrifuge at 400 × g for 5 min at RT.
7. Gently remove supernatant, cells are ready for further proces-
sing or other analysis. The purified PGCs isolated from the
bloodstream or gonads are presented in Fig. 4.
3.4.2 Identification of
PGCs
1. Fix purified 10 μL suspension PGCs in 10 μL 4% glutaralde-
hyde for 5 min to dry.
PAS Staining
2. Rinse twice in 1 × PBS.
3. Immerse cells in a 0.5% aqueous periodic acid solution for
5 min at room temperature.
4. Wash samples twice with 1 × PBS and incubate with Schiff’s
Reagent for 15 min at RT.
5. Wash samples twice with 1 × PBS and observed under a light
microscope (Fig. 5).
35. Purification and Transfection of PGCs 21
Fig. 3 Percoll gradient with a layer of cells isolated from the bloodstream of
embryos at 14–16 HH contained suspension of bPGCs and blood counts (arrow).
The bottom layer (50% Percoll solution) was stained by phenol red
Immunocytochemistry 1. Take 100 μL purified cells (approx. 4.5 × 105
cells) and add
1 mL 1 × PBS, centrifuge at 400 × g for 5 min, and resuspend
in 1 × PBS to rinse culture medium.
2. The PGCs suspension fix in 1% paraformaldehyde for 15 min.
3. Wash in 1 × PBS and centrifuge at 400 × g for 5 min.
4. Block cells with 2.5% goat serum for 30 min at RT.
5. Centrifuge at 400 × g for 5 min.
36. 22 Luiza Chojnacka-Puchta and Dorota Sawicka
Fig. 4 The PGCs isolated from chicken embryos and purified via Percoll centrifugation density. PGCs are round
cells ranging in diameter from 10 and 14 μM with a large nucleus and large, clearly visible vacuoles. (a) bPGCs
(20×) and (b) gPGCs (40×)
Fig. 5 The purified chicken PGCs stained with PAS. Positive results of staining (red- stained cells) indicate the
presence of glycogen vesicles in the PGCs, which confirms their origin. (a) bPGCs (20×) and (b) gPGCs (40×)
6. Incubate cells with an antibody against a specific PGC marker
(anti-stage-specific embryonic antigen 1 (SSEA-1)-FITC/
PerCP) for 1 h according to the manufacturer’s protocol (see
Note 6).
7. Observe samples under fluorescent microscope at an excitation
wavelength of 488 nM using a 515/30 filter or 682/33
(respectively for used fluorochromes) (Fig. 6).
3.5 Transfection
Methods and Culture
of PGCs
1. Centrifuge purified PGCs at 300 × g for 3 min and resuspend in
medium without any additions (0.45 mL).
2. Add plasmid (20 μg) and transfer cells to 0.4 mM cuvette.
3.5.1 Electroporation 3. Perform electroporation under conditions: 200 V, 900 μF.
4. Transfer cells into the well of a 4-well plate and culture for 24 h
(37 °C and 5%, CO2).
5. After 24 h, replace the medium with a culture complex medium
(see Notes 7 and 8).
37. Purification and Transfection of PGCs 23
Fig. 6 The purified chicken PGCs subjected to immunofluorescence staining with antibodies for the specific
cell surface antigen SSEA-1 after excitation wavelength of 488 nM. Staining of PGCs in green by anti-SSEA1-
FITC (a) and red by anti-SSEA1-PerCp (b) confirmed their identity (20×)
3.5.2 Lipofection 1. Seed isolated and purified PGCs into 4-well plate.
2. All reagents: X-tremeGENE HP DNA Transfection Reagent,
DNA and medium must be at RT (15–25 °C) (see Note 9).
3. Dilute DNA with medium OptiMEM I without any additions
to a final concentration of 1 μg plasmid DNA/100 μL medium.
4. Transfer 100 μL of medium containing 1 μg DNA into a
sterile tube.
5. Briefly vortex vial of Transfection Reagent and add 3 μL
directly into the medium containing DNA from step 3. Mix
gently.
6. Incubate for 15 min at RT.
7. Add transfection complex to PGCs in a dropwise manner.
Gently shake the wells.
8. Incubate cells for 24 h.
9. Change medium to culture complex medium (see Notes 7
and 8).
4 Notes
1. Prepare the incubator with conditions: 37.8 °C and 60–62%
relative humidity. At least 50 eggs for one isolation is needed.
The most important thing is the temperature in the egg incu-
bator. Plug in and turn on the incubator. Wait for the tempera-
ture to stabilize. Remember, the more eggs you have, the
longer it will take to get to the temperature and stabilize.
Because the warm air naturally rises to equalize the warm air
in each part of the incubator, install a fan with air circulation
38. 24 Luiza Chojnacka-Puchta and Dorota Sawicka
best. Also, we recommend using a hygrometer, a device that
measures humidity in the air. To increase the humidity, put a
container with distilled water into the incubator.
2. Place the eggs in a horizontal position and leave for 5–10 min,
allowing the embryo to assume the appropriate position when
it is released from the egg.
3. 100% Percoll stored at 4 °C should have pH 8.5–9.5. We find
that Percoll solutions is best to prepare fresh each time, before
density gradient centrifugation. They need to be used at room
temperature. Use phenol red to provide visual contrast for
different phases. Add drop-by-drop directly to each 3 mL of
load density successively: 50%, 25% and 12.5% into clear 15 mL
tube, avoiding bubble formation.
4. Carefully layer cells onto the top of the 12.5% layer immediately
before centrifugation. Place the gradient into a swinging
bucket rotor carefully and centrifuge.
5. Carefully remove the gradient and collect PGCs from above the
red layer between 50 and 25 phases. To make collection easier,
first, remove the top layer.
6. We used an antibody sc-21702 Santa Cruz Biotechnology,
Santa Cruz, CA, USA.
7. Change medium every 3 days and culture cells prior to further
use (injection or cryoconservation). We recommend using
OptiMEM I medium with supplements: 2% CS, 10% FBS,
20 ng/mL bFGF, 9 ng/mL mLIF, 5 ng/mL hSCF, 1% antibi-
otic and selective antibiotic.
8. Check the effectiveness of electroporation PGCs based on
viability if you use selective antibiotics or other markers. Add
selective antibiotic after transfection to obtain stable transfec-
tants. We used genetycin (G418) at a concentration of
50 μg/mL.
9. Perform lipofection of PGCs with the use of one of the com-
mercially available lipofectants. In our laboratory we use the
X-tremeGENE HP DNA Transfection Reagent (Roche) and
lipofection protocol according to the manufacturer’s instruc-
tions. We recommend the use of a 3:1 rate. This procedure
does not require the removal of lipofectant (e.g.,
X-tremeGENE HP DNA Transfection Reagent or FUGENE
6 Transfection Reagent), which may be needed for the other
reagents.
Acknowledgments
This paper was partially supported by resources from the Ministry
of Education and Science and also funds intended for the
39. maintenance and progress of research with didactic potential. Task
no. I-61. This work was also partially supported by the National
Science Center (NCN 2018/02/X/NZ9/00997 2018).
Purification and Transfection of PGCs 25
References
1. van de Lavoir MC, Diamond JH, Leighton PA,
Mather-Love C, Heyer BS, Brashaw R,
Kerchner A, Hooi LT, Gessaro TM, Swanberg
SE, Delany ME, Etches RJ (2006) Germline
transmission of genetically modified primordial
cells. Nature 441:766–769
2. Eyal-Giladi H, Kochav S (1976) From cleavage
to primitive streak formation: a complementary
normal table and new look at the first stages of
the development of the chick. I. General mor-
phology. Dev Biol 49:321–337
3. Hamburger V, Hamilton HL (1951) A series of
normal stages in the development of the chick
embryo. J Morphol 88:49–92
4. Naito M, Sano A, Harumi T, Matsubara Y,
Kuwana T (2004) Migration of primordial
germ cells isolated from embryonic blood into
the gonads after transfer to stage X blasto-
derms and detection of germline chimaerism
by PCR. Br Poult Sci 45:762–768
5. Chojnacka-Puchta L, Sawicka D, Lakota P,
Plucienniczak G, Bednarczyk M, Plucienniczak
A (2015) Obtaining chicken primordial germ
cells used for gene transfer: in vitro and in vivo
results. J Appl Genet 56(4):493–504
6. Sawicka D, Chojnacka-Puchta L, Zielinski M,
Plucienniczak G, Plucienniczak A, Bednarczyk
M (2015) Flow cytometric analysis of apoptosis
in cryoconserved chicken primordial germ
cells. Cell Mol Biol Lett 20(1):143–159
7. Zhao DF, Kuwana T (2003) Purification of
avian circulating primordial germ cells by nyco-
denz density gradient centrifugation. Br Poult
Sci 44:30–35
8. Park TS, Jeong DK, Kim JN, Song GH, Hong
YH, Lim JM, Han JY (2003) Improved germ-
line transmission in chicken chimeras produced
by transplantation of gonadal primordial germ
cells into recipient embryos. Biol Reprod 68:
1657–1662
9. Oishi I (2010) Improvement of transfection
efficiency in cultured chicken primordial germ
cells by percoll density gradient centrifugation.
Biosci Biotechnol Biochem 74(12):
2426–2430
10. Yamamoto Y, Usui F, Nakamura Y, Ito Y,
Tagami T, Nirasawa K, Matsubara Y, Ono T,
Kagami H (2007) A novel method to isolate
primordial germ cells and its use for the gener-
ation of germline chimeras in chicken. Biol
Reprod 77(1):115–119
11. Mozdziak PE, Angerman-Stewart J,
Rushton B, Pardue SL, Petitte JN (2005) Iso-
lation of chicken primordial germ cells using
fluorescence-activated cell sorting. Poult Sci
84:594–600
12. Ginsburg M, Eyal-Giladi H (1986) Temporal
and spatial aspects of the gradual migration of
primordial germ cells from the epiblast into the
germinal crescent in the avian embryo. J
Embryol Exp Morphol 95:53–71
13. Meyer DB (1960) Aplication of the periodic
acid–Schiff technique to whole chick embryos.
Biotech Histochem 35:83–89
14. Macdonald J, Glover JD, Taylor L, Sang HM,
McGrew MJ (2010) Characterisation and
germline transmission of cultured avian pri-
mordial germ cells. PLoS One 5:e15518
15. Quaye Mensah B (2023) Ligamentum arterio-
sum and its telocytes: an ultrastructure descrip-
tion. Anat Rec (Hoboken) 306(1):187–192.
https:/
/doi.org/10.1002/ar.25052. Epub
2022 Aug 6. PMID: 35904054
16. Resnick JL, Bixler LS, Cheng L, Donovan PJ
(1992) Long-term proliferation of mouse pri-
mordial germ cells in culture. Nature 359:550–
551
17. Urven LE, Erickson CA, Abbott UK, McCar-
rey JR (1988) Analysis of germ line develop-
ment in the chick embryo using an anti-mouse
EC cell antibody. Development 103:299–304
18. Karagenc L, Cinnamon Y, Ginsburg M, Petitte
JN (1996) Origin of primordial germ cells in
the prestreak chick embryo. Dev Genet 19:
290–301
19. Tsunekawa N, Naito M, Sakai Y, Nishida T,
Noce T (2000) Isolation of chicken vasa
homolog gene and tracing the origin of pri-
mordial germ cells. Development 127:2741–
2750
41. may become azoospermic [3–5]. Consequently, current research
efforts have focused on SSC therapy to treat male factor infertility.
28 Janmejay Hingu et al.
Spermatogenesis begins with the SSC, a diploid cell that main-
tains the ability for self-renewal and downstream differentiation
into spermatozoa. Given its origin in spermatogenesis, using SSC
for male infertility is a logical choice for patients with maturation
arrest or initially diagnosed SCO with salvageable SSC on the
biopsy [2, 6]. Investigative SSC therapies have included SSC trans-
plantation and in vitro spermatogenesis. The formed differentiated
germ cells could potentially be used for in vitro fertilization (IVF),
either intracytoplasmic sperm injection (ICSI) or experimental
round spermatid injection (ROSI) [7–10]. Given that SSC is a
scarce population in azoospermic patients, in vitro propagation of
SSC is a critical step toward the feasibility of stem cell-based fertility
treatments.
This chapter aims to provide our institution’s protocol and
technical considerations for SSC culture in mammalians. Previous
works from our lab have included SSC isolation, propagation, and
partial differentiation to the round spermatid level in a unique
culture system [10–12]. This was initially performed in adult men
where SSC could be isolated and propagated for 28 weeks up to a
concentration of 18,540-fold [11]. Subsequently, this application
was expanded to prepubertal boys in which SSCs were cultured for
up to 29 weeks [12]. Most recently, this culture system has been
successfully applied to a mouse model and human patients with
Klinefelter syndrome, given the frequency of azoospermia, negative
mTESE, and the need for alternative therapies [8, 9, 13].
2 Materials
2.1 Reagents 1. Phosphate-buffered saline (PBS, 1×).
2. Minimal Essential Media 1×.
3. Fetal calf serum.
4. DNase (Roche) stock solution of 2 mg/mL. It is stored at -
20 °C in 200 μL aliquots. Keep in ice before use.
5. Collagenase NB 4 (Serva) stock solution of 10 PZ U/mL. It is
stored at -20 °C in 200 μL aliquots. Keep in ice before use.
6. Neutral Protease NB (Serva) stock solution of 10 DMC
U/mL. It is stored at -20 °C in 100 μL aliquots. Keep in ice
before use.
7. Enzyme mix: 5 mL 1× MEM DNase (8 μg/mL) + SERVA
Collagenase 4 PZ U + neutral Protease* 0.2 DMC
U + 20 μL extra DNase (2 mg/mL). If cells are intended for
transplantation into humans, Neutral Protease NB GMP Grade
is recommended.
42. Isolation and In Vitro Propagation of Human Spermatogonial Stem Cells 29
8. Trypsin-EDTA 0.25% (Gibco).
9. 1 × MEM 10%FBS.
10. StemPro™ complete medium (Tables 1 and 2) (see Note 1).
2.2 Equipment 1. Laminar flow cabinet.
2. Shaking water bath with temperature control.
3. Water bath or bean bath with temperature control.
4. Centrifuge.
5. Microcentrifuge.
6. Pipette controller.
7. Serological pipettes (5, 10 mL).
8. Micropipettes.
9. Micropipette tips (2–1000 μL).
10. Conical tubes (10, 50 mL).
11. Microcentrifuge tubes (1.5 mL).
12. Nylon filter (77 μM, 55 μM).
13. Culture plates (size and surface will depend on the cells
obtained).
14. Petri dishes.
15. Gloves.
16. Tweezers.
17. Surgical blade.
18. Aluminum foil.
19. Parafilm.
20. Ice.
21. Cell culture incubator.
22. Optical microscope.
23. Dissection microscope or stereoscope.
24. Hemocytometer or Bürker-Turk cell counting slide.
3 Methods
3.1 Before Isolation 1. Preheat water or bean bath to 37 °C. Fill with autoclaved water.
Set culture media within to incubate before use.
2. Preheat the shaking water bath to 32 °C. Fill with autoclaved
water.
3. Ensure sterile conditions are present inside the laminar flow
cabinet.
43. Table
1
Chemicals
used
to
supplement
StemPro-34™
SFM
–
–
–
–
–
–
-
–
–
–
–
–
–
–
–
L
5
-
–
–
L
1
–
–
–
–
L
30
nM
L
60
–
–
30 Janmejay Hingu et al.
Chemicals/media
Storage
Stock
conc.
Solvent
Amount
for
500
mL
Final
conc.
Solids
Bovine
albumin
4
°C
2.5
g
5
mg/mL
D(+)
glucose
RT
3
g
6
mg/mL
Ascorbic
acid
RT
8.8
mg
1
×
10
4
M
Apo-Transferrin
-20
°C
50
mg
100
μg/mL
Pyruvic
acid
(sodium
pyruvate)
4
°C
15
mg
30
mg/mL
D-Biotin
4
°C
5
mg
10
μg/mL
Liquids
2-betamercaptoethanol
RT
1.7
μ
×
10
5
M
DL-lactic
acid
4
°C
500
μ
μL/mL
MEM
non-essential
amino
acids
4
°C
5
mL
10
μL/mL
StemPro™-34
SFM
Nutrient
Supplement
-20
°C
13
mL
26
μL/mL
Insulin
-20
°C
12.5
mg/mL
10
mM
HCl
1
mL
25
μg/mL
Sodium
selenite
-20
°C
0.25
mg/mL
milliQ
water
10
μ
Putrescine
-20
°C
100
mg/mL
milliQ
water
50
μ
μM
L-glutamine
-20
°C
200
mM
–
5
mL
2
mM
MEM
gitamin
solution
-20
°C
5
mL
10
μL/mL
b-Estradiol
-20
°C
0.6
mg/mL
100%
EtOH
25
μL
30
ng/mL
Progesterone
-20
°C
0.6
mg/mL
100%
EtOH
50
μL
60
ng/mL
RT
stands
at
room
temperature
45. 32 Janmejay Hingu et al.
4. Ensure that the incubator has achieved optimal conditions for
culture (37 °C, 5% CO2) and shows no signs of contamination.
5. The sample of testis tissue can be received from the operating
room freshly or already cryopreserved viable tissue. The isola-
tion process may vary slightly depending on whether fresh or
frozen testis tissue is used.
6. Prepare 1× MEM with DNase mix (20 μL of 2 mg/mL DNase
per 5 mL 1× MEM). Each sample will require about 100 mL.
7. Prepare 1× MEM 10% FBS (without DNase) for cell culture.
8. Prepare StemProTM
Complete for cell culture (Tables 1 and 2)
(see Note 2).
3.2 Isolation 1. Transfer approximately 20 mL of 1× MEM/DNase into three
Petri dishes.
2. If the sample is fresh, deposit it on a petri dish with 1× MEM/-
DNase and move on to step 5. If the tissue is frozen, allow it to
adjust to room temperature for 2 min. Then continue thawing
the cryovial with lukewarm running tap water and wipe off
residual water with 70% ethanol (see Note 3).
3. As thawing continues, the tissue inside the cryotube will grad-
ually transition from solid to liquid. When the tissue becomes
liquid enough, but there is still a portion of solid pellet yet to be
thawed, empty the cryovial contents into a petri dish contain-
ing 1× MEM/DNase for the first wash.
4. Transfer the tissue into a second fresh petri dish containing 1×
MEM/DNase using a pair of tweezers. Next, move the tissue
into a new petri dish (the third one) containing 1× MEM/-
DNase (see Note 4).
5. Remove the tunica albuginea, if necessary, using a pair of
tweezers and a surgical blade (see Note 5).
6. Separate the seminiferous tubules of the testis apart and remove
connective tissue, if present, using tweezers under a dissection
microscope (Fig. 1). Note the tubules’ appearance (thick, thin,
long, short, etc.) (see Note 6).
Fig. 1 Bright-field microscopic view of seminiferous tubules before the addition of enzyme mix (a, b) and
subsequent incubation in which the lumens of the tubules appear opaque (a, b) and transparent (c)
46. Isolation and In Vitro Propagation of Human Spermatogonial Stem Cells 33
7. Transfer the tissue suspended in 1× MEM/DNase to a
15 mL tube.
If tube contents appear clear, allow tubules to settle at the
bottom of the tube. If tube contents appear turbid, centrifuge
(5 min at 65 g without brake), and remove the supernatant.
Wash with 1× MEM/DNase until the supernatant is clear.
8. Remove the supernatant, if present, from the tube until only
5 mL are left. Then add 5 mL of Enzyme Mix, bringing the
total volume to 10 mL.
9. Close the tube and seal the cap with parafilm. Submerge the
tube completely in the shaking water bath and incubate for
30 min (32 °C, 120 cycles/min).
10. Remove the tube from the incubator and open it. Without
pipetting up and down, take a 10 μL sample and place it on a
microscope slide for viewing (Fig. 1). If present, note the
thickness of the peritubular cell layer and any fenestrations.
Incubate longer, if necessary, until the tubules appear fene-
strated, with most extra tubular cells released but still full of
intratubular cells inside.
11. Centrifuge the tube (5 min at 16 g) without brake. Remove the
supernatant if present.
12. Wash with 5 mL 1× MEM/DNase. Centrifuge (5 min at 16 g),
without brake, and discard the supernatant.
13. Add 5 mL of fresh Enzyme Mix.
14. Close the tube and seal the cap with parafilm. Submerge the
tube completely in the shaking water bath and incubate for
30 min (32 °C, 120 cycles/min).
15. Remove the tube from the incubator and open it. Pipette
gently up and down about ten times. Take 10 μL and place it
on a microscope slide for viewing (Fig. 1). Note changes in the
peritubular cell layer thickness and fenestrations since the first
incubation (see Note 7).
16. Discard any clumps of tubules, if present.
17. Filter the entire suspension through a 70 μM nylon filter into a
new 50 mL tube. Then, filter the rest again through a 40 μM
nylon filter into a new 50 mL tube. Cells of the exact origin
may be pooled together during filtration.
18. Centrifuge (5 min at 350 g without brake).
(a) If the supernatant is clear, discard it.
(b) If the supernatant is turbid, take 10 μL and place it on a
microscope slide for viewing. If large cells are present,
repeat centrifugation to avoid losing stem cells.
47. 34 Janmejay Hingu et al.
Fig. 2 Isolated testicular cells in culture. Seeded single cells after isolation (a), Germline Stem Cell cluster
formation on top of somatic cells 3 weeks after cell culture (b, c)
19. Add 500 μL 1× MEM to the pellet and resuspend gently.
Transfer to a microcentrifuge tube and spin down (5 min at
180 g, with brake).
20. Discard the supernatant and dissolve the pellet in ±500 μL 1×
MEM/10% FCS (without DNase), depending on the size of
the cell pellet.
21. Count the isolated cells using a Hemocytometer.
22. Culture cells in a 6-well plate with 1× MEM/10% FBS at a
concentration of 5000–10,000 cells/cm2
. Incubate overnight
at 37 °C, 5% CO2.
3.3 Cell Culture 1. Keep the cells inside the incubate at 37 °C, 5% CO2, replacing
1× MEM/10% FBS with StemPro complete medium (Tables 1
and 2).
2. Observe the cells in the culture regularly (Fig. 2). Note cell
morphology, the confluence of attached cells, and cell debris or
contamination (see Notes 8 and 9).
3. Refresh culture media 2–3 times a week. Use the media volume
recommended by the manufacturer. Avoid losing floating cells
by collecting spent media in a tube and centrifuging to form a
pellet. Resuspend the pellet with new media and reintroduce
the volume evenly among culture wells. Shake the plate gently
to ensure the cells seeded spread evenly around the well (see
Note 10).
4. When 80–90% confluency is achieved, cells should be trypsi-
nized and passage into a new plate to continue propagation. If
somatic cell overgrowth is suspected, differential plating can be
considered 12–48 h after seeding.
3.4 Cell
Cryopreservation
1. Passage cells with trypsin EDTA incubation and then
count them.
2. Plan the way you want to freeze your cells depending on the
study’s goals and cell availability (see Note 11).
48. Isolation and In Vitro Propagation of Human Spermatogonial Stem Cells 35
3. Centrifuge cells (5 min, 180 g, with break). Remove the super-
natant and resuspend the pellet in 750 μL 1× MEM/20% FCS
(Room Temperature).
4. Transfer the cell suspension into a cryovial.
5. Add 750 μL 1× MEM/20% FCS/16% DMSO while gently
shaking the cryovial.
6. Transfer the cryovial into a cold Nalgene freezing box
(Mr. Frosty). Maintain at -80 °C for around 12 h but no
longer than 24 h.
7. Transfer the cryovial to a liquid N2 tank.
4 Notes
1. The reagents shown in Table 1 are added to 500 mL
StemPro™-34 serum-free medium to supplement the
medium. Final concentrations are based on the 500 mL
StemPro™-34 SFM, not the total volume.
2. After mixing reagents, filter the mixture with vacuum pressure
over a 22 μM filter. Immediately before use, add growth factors
shown in Table 2 to 50 mL supplemented StemPro™ SFM.
3. An aggressive change in the temperature of the cryovial may
trigger an expansive reaction of the gas inside. This may break
the cryovials, compromising the sample integrity or even
becoming a safety hazard for the operator. Ensure the initial
thawing is gradual and that appropriate safety guideline is
followed. We also recommend briefly opening the cryovials to
release air pressure as soon as the thawing process allows.
4. As previously reported, dimethyl sulfoxide DMSO is one of the
most commonly used cryoprotectants for testicular tissue [14],
but at room temperature, DMSO becomes cytotoxic. Three
time-sensitive consecutive washes are done to minimize tissue
exposure to DMSO at room temperature.
5. During this step, it is recommended to weigh the tissue to
calculate the isolation efficacy (number of cells per mg tissue).
6. Seminiferous tubules from simple testicular biopsies are usually
more laborious to separate than in mTESE samples. The
further separated the tubules are, the more adherent
(mucous-like) they appear and the more influential the enzy-
matic digestion will be.
7. The tubular cell layer should disintegrate at this point, releasing
the stem cells. The thickness of the tubular cell layer can differ
between patients. The incubation with enzymes can be
continued for longer to achieve the desired characteristics of
the peritubular cell layer. If cell clumps form, dissociate them
with trypsin-EDTA.
49. 8.
36 Janmejay Hingu et al.
8. Taking pictures of the cells in culture is encouraged.
9. If there is uncertainty about cell viability, Live/Dead staining
(Invitrogen LIVE/DEAD™ Viability/Cytotoxicity Kit) fol-
lowed by fluorescence microscopy can be performed before
refreshing media.
10. Avoid keeping cultured cells without media for too long dur-
ing the refresh process.
11. In our experience, we use 1.8 mL cryovials with up to
4,000,000 cells per cryovial. Higher cell concentrations may
compromise cell viability after thawing.
References
1. Zegers-Hochschild F et al (2017) The interna-
tional glossary on infertility and fertility care,
2017. Hum Reprod 32(9):1786–1801
2. Abdelaal NE et al (2021) Cellular therapy via
spermatogonial stem cells for treating impaired
spermatogenesis, non-obstructive azoosper-
mia. Cell 10(7):1779
3. Halpern JA et al (2020) Oncofertility in adult
and pediatric populations: options and barriers.
Transl Androl Urol 9(Suppl 2):S227–S238
4. David S, Orwig KE (2020) Spermatogonial
stem cell culture in oncofertility. Urol Clin
North Am 47(2):227–244
5. Diao L et al (2022) Roles of spermatogonial
stem cells in spermatogenesis and fertility res-
toration. Front Endocrinol (Lausanne) 13:
895528
6. Xi HM et al (2022) Recent advances in isola-
tion, identification, and culture of mammalian
spermatogonial stem cells. Asian J Androl
24(1):5–14
7. Tanaka A et al (2018) Ninety babies born after
round spermatid injection into oocytes: survey
of their development from fertilization to
2 years of age. Fertil Steril 110(3):443–451
Galdon G et al (2021) In vitro propagation of
XXY undifferentiated mouse spermatogonia:
model for fertility preservation in Klinefelter
syndrome patients. Int J Mol Sci 23(1):173
9. Galdon G et al (2022) In vitro propagation of
XXY human Klinefelter spermatogonial stem
cells: a step towards new fertility opportunities.
Front Endocrinol (Lausanne) 13:1002279
10. Pendergraft SS et al (2017) Three-dimensional
testicular organoid: a novel tool for the study
of human spermatogenesis and gonadotoxicity
in vitro. Biol Reprod 96(3):720–732
11. Sadri-Ardekani H et al (2009) Propagation of
human spermatogonial stem cells in vitro.
JAMA 302(19):2127–2134
12. Sadri-Ardekani H et al (2011) In vitro propa-
gation of human prepubertal spermatogonial
stem cells. JAMA 305(23):2416–2418
13. Deebel NA et al (2022) Morphometric and
immunohistochemical analysis as a method to
identify undifferentiated spermatogonial cells
in adult subjects with Klinefelter syndrome: a
cohort study. Fertil Steril 118(5):864–873
14. Zarandi NP et al (2018) Cryostorage of imma-
ture and mature human testis tissue to preserve
spermatogonial stem cells (SSCs): a systematic
review of current experiences toward clinical
applications. Stem Cells Cloning 11:23–38
51. possibility to isolate and analyze male germ cells at the defined stage
of differentiation. In this regard, the STA-PUT technique repre-
sents, since the late 1960s, a pivotal procedure in this field of study.
This technique takes advantage of the fact that the individual types
of testicular cells have different size, thus sedimenting at different
velocities in a BSA gradient, independently from the shape. The
physical characteristics of the method have been extensively
described by Miller and Phillips using blood cells [8] and then
used for the separation of mouse male germ cells in the laboratory
of Dr. W.R. Bruce [9]. The effectiveness of the technique has been
re-evaluated from a morphological point of view [10], re-examined
also recently proposing different equipment [11], and widely used
for the study of DNA synthesis and transcriptional and translational
activity in the different phases of spermatogenesis and for a variety
of biochemical and metabolic studies [12–16]. The recent advance-
ment of transcriptomic approaches has provided remarkable
insights into the transcriptional and post-transcriptional molecular
mechanisms underlying the tightly and dynamically controlled gene
programs promoting male germ cells differentiation [17, 18]. Most
of these studies exploited RNA-sequencing (RNA-seq) from either
bulk-testis, collected at different time-points during the first syn-
chronous wave of murine spermatogenesis, or from male germ cells
at different stage of differentiation, isolated by either centrifugal
elutriation or Fluorescence-activated Cell Sorting (FACS) [3, 6,
7]. Both these approaches show limitations: i. presence of hetero-
geneous populations of somatic cells (i.e., Sertoli cells, Leydig cells)
is a confounding element in the identification of germ-cell-specific
gene expression patterns by RNA-seq of bulk-testes; ii. stage-
specific male germ cell isolation by centrifugal elutriation requires
injection of a high number of testicular cells obtained by pooling
testes from multiple mice. This latter aspect is a critical limitation
for transcriptomic studies aimed at the identification of gene
expression alterations correlated to spermatogenic defects in mice
with reduced reproductive potential, due to the small number of
animals. In our laboratory, we showed that gravimetric decantation
(STA-PUT) of testicular cellular suspension is a reliable method to
collect homogenous population of meiotic (spermatocytes) and
post-meiotic (round spermatids) male germ cells from a single
mouse, from which it is possible to isolate a sufficient amount of
RNA for reliable total-RNA seq analysis [4]. Moreover, we showed
this method to be applicable and reliable also for the isolation of
germ cells and subsequent transcriptomic analysis from a mouse
model with relevant spermatogenic defects, with reduced testis size
and impaired male germ cells differentiation, such as the Sam68
knock-out model [19, 20], therefore proving its applicability to the
late-generation transcriptomic analysis. Herein, we will describe a
detailed protocol for the fractionation of meiotic spermatocytes
and post-meiotic round spermatids from a single adult mouse and
the isolation of good-quality RNA for further transcriptomic
analysis.
38 Chiara Naro et al.
52. Male Germ Cells Isolation from Single Mouse Using STA-PUT 39
2 Materials
2.1 Animals The procedure described in this chapter refers to testicular germ
cells suspension obtained from the digestion of testes of 12-week
old mouse from the C57BL/6 strain (see Note 1).
2.2 BSA Gradients 1. 1.5 L EKRB (120.1 mM NaCl, 4.8 mM KCl, 25.2 mM
NaHCO3, 1.2 mM KH2PO4, 1.2 mM MgSO4, 1.3 mM
CaCl2, 11 mM). All components are diluted in water. This
solution is used for the preparation of both the BSA gradients
and the testicular germ cells suspension. Solution is filtered
through a 0.22 μM filter and stored at 4 °C (see Note 2).
2. Bovine serum albumin (BSA) 3% in EKRB: Dissolve 12 g BSA
in 400 mL of EKRB at room temperature using magnetic
stirring. The solution is filtered through a 0.22 μM filter and
stored at 4 °C. 250 mL and will be used as such to create the
gradient. The excess will be used to prepare 250 mL of 1% BSA
in EKRB, 60 mL 0.5% BSA in EKRB, and 15 mL 0.2% BSA in
EKRB by the dilution of an appropriate amount of EKRB w/o
BSA (see Notes 3 and 4).
2.3 STA-PUT
Apparatus
1. Sedimentation chamber: The sedimentation chamber had a
conical base carved out of a square plate with a side of 15 cm
and a thickness of 4 cm. The cone had a diameter of 12 cm at
the base and was 3 cm deep with a hole of 4 mm diameter at the
apex. Above the conical base a 15 cm long tube with an internal
diameter of 12 cm was glued to complete the chamber.
2. Three cylinders, necessary for the formation of the albumin
gradient to be loaded into the sedimentation chamber, with the
following characteristics:
(a) Two cylinders measuring 7.5 cm internal diameter and
15 cm height on a square base with 10 cm sides to ensure
stability. At the base of one cylinder (n.1) there is one
outlet represented by a small conical plexiglass tube with
an internal diameter of 2 mm; at the base of the second
cylinder (n.2), there are two of such outlets placed at the
two ends of the cylinder diameter.
(b) The third cylinder measured 1.8 cm internal diameter and
15 cm height on a square base of 5 cm sides to ensure
stability. At the base of the cylinder (n.3), there are two
outlets as described for cylinder n.2 (see Note 5).
3. One baffle: A 1.2 cm diameter stainless steel hemisphere with
three 2 mm pegs (see Note 6).
4. Silicone tubing 2 mm internal diameter.
53. 40 Chiara Naro et al.
5. Straight mini tubing connector to fix the tubing to the base
hole of the STA-PUT.
6. One 10 mL syringe for sample injection, hold by a support
clamp, attached through a bosshead to a laboratory stand.
7. Two 3-way stopcocks and luer lock adapters if necessary to
connect tubing.
8. Three curved tubing clamps, placed on the connecting tubes
between the cylinders and the syringe.
9. One tubing clip, Hoffman pattern, placed on the connecting
tube between the syringe for sample injection and the STA-
PUT chamber.
10. Two magnetic stirrers for cylinders n. 2 and 3.
11. Two magnetic stirring bars for cylinders n. 2 and 3.
12. A small laboratory jack to support the cylinder n.1 at the same
level of cylinders 2 and 3.
13. A perforated support about 10 cm high for the STA-PUT
(laboratory jack or polystyrene box).
2.4 Testicular Germ
Cells Preparation
1. Shaking water bath set at 32 °C.
2. Refrigerated bench centrifuge.
3. Sterile blades.
4. 15 mL sterile polypropylene tubes.
5. Hemocytometer for cell counting.
6. Minimum Essential Medium (MEM).
7. Collagenase from Clostridium histolyticum (Sigma Aldrich,
Cat # C7657; CAS 9001-12-1): 5 mg/mL in MEM, equal to
a 20-fold stock solution (see Note 7).
8. DNase I from bovine pancreas (Sigma Aldrich, Cat # DN25;
CAS 9003-98-9): 2 mg/mL dissolved in MEM, equal to a
40-fold stock solution (see Note 7).
9. Trypsin from bovine pancreas (Sigma Aldrich Cat # T9201;
CAS 9002-07-7): 10 mg/mL in MEM, equal to a ten-fold
stock solution (see Note 7).
10. Fetal bovin serum (FBS).
2.5 Fractions
Collection and
Analysis of Germ Cells
Nuclear Morphology
1. Refrigerated bench centrifuge.
2. 15 and 50 mL sterile polypropylene tubes.
3. 1.5 mL sterile polypropylene tubes.
4. Hoechst 33342 10 mg/mL in water (ThermoFisher Scientific
Cat #H3570).
5. Microscope slides.
54. Male Germ Cells Isolation from Single Mouse Using STA-PUT 41
2.6 RNA Purification 1. Refrigerated bench centrifuge.
2. miRNeasy Mini Kit with DNase (Qiagen).
3. DNAse-RNAse free 1.5 mL tubes and tips (see Note 8).
3 Methods
3.1 Assembly of the
STA-PUT Apparatus
Prior or during the germ cells isolation procedure, place the appa-
ratus in a cold room or in a large refrigerator, where all procedures
will be performed. The setup of the apparatus is illustrated in Fig. 1.
1. Place Cylinders n. 2 and n. 3, with a stirring bar of the proper
size inside, on two magnetic stirrers. Place the cylinder n. 1 on a
laboratory jack of the same height as the stirrers. The three
cylinders should be connected with silicone tubing and placed
on a shelf approximately 30 cm above the base of the STA-PUT
to allow for gradient flow during loading.
2. Place firmly the STA-PUT chamber, with the baffle inside (see
Note 9), on the perforated support to allow the passage of the
cell suspension and the gradient to and from the base hole of
the chamber.
Fig. 1 The STA-PUT apparatus: (a) cylinder n.1 containing 3% BSA; (b) cylinder
2 containing 1%BSA; (c) cylinder 3 containing 5% BSA; (d) 10 mL syringe for
EKRB and cell suspension loading; (e) silicone tubing; (f) magnetic stirring bars;
(g) curved tubing clamps; (h) three-way stopcock; (i) small laboratory jack; (l)
magnetic stirrer; (m) laboratory stand; (n) support clamp and boss-head; (o)
tubing clips, Hoffman pattern; (p) perforated laboratory jack; (q) STA-PUT; (r)
baffle; (s) straight mini tubing connector; (t) exit for the collection of fractions
56. at .28.
Sold to J. C. Peters Son, 267 Roberts St., 3 chests Oolong tea, 146# at
.52, 5 sacks Rio coffee, 252# at .48.
Bought for cash from Harris Co., 1 office desk and chair $45.00, gave
Ck. No. 3 in payment.
—4th—
Paid freight on coffee from New York by Ck. No. 4, 12.93.
Sold to Wright Noble, 146 7th St., 2 sacks Java coffee, 99# at .32; 2
sacks Mocha coffee, 101# at .32.
Sold to Horgis Co., 84 Jackson St., 5 chests Japan tea, 248# at .48.
Sold to Winters James, 92 Hastings St., 4 chests Japan tea, 201# at
.48; 3 chests Oolong tea, 138# at .52; 2 sacks Java coffee, 97# at .32.
Sold for cash 1 sack Rio coffee, 47# at .28; 1 chest Japan tea, 45# at
.48.
—5th—
Sold to Cobb Willet, 892 Park Av., 2 chests Japan tea, 92# at .48; 1
chest Oolong tea, 47# at .52; 1 sack Rio coffee, 44# at .28; 1 sack Java
coffee, 45# at .32; 1 sack Mocha coffee, 43# at .32.
Sold to Young Criger, 62 Watson St., 5 sacks Mocha coffee, 205# at
.32; 3 chests Oolong tea, 127# at .52.
Bought from Japan Importing Co., San Francisco, 60 chests Japan tea,
2,700# at .36, f. o. b. Omaha, net cash; gave our note at 10 days
without interest in payment.
Paid account of Leggitt Co., less 2% discount, Ck. No. 5.
—6th—
Ames Johnson paid their account, less 2% cash discount.
Deposited cash received to date.
Sold to Wade Francis, 92 Bluff St., 10 chests Japan tea, 448# at .48.
Paid for telegram—petty cash—.40.
57. Received check from Landis Snow in full settlement of their account.
Sold to J. C. Peters Son, 5 sacks Java coffee, 231# at .32.
Sold for cash 3 sacks Rio coffee, 127# at .28; 2 sacks Mocha coffee,
89# at .32; 3 sacks Japan tea, 131# at .48.
—8th—
Deposited cash on hand, also check of Landis Snow.
Sold to Ames Johnson, 2 sacks Mocha coffee, 91# at .32; 2 chests
Oolong tea, 87# at .52.
Sold to Wright Noble, 3 chests Japan tea, 129# at .48; 1 chest Oolong
tea, 42# at .52.
Paid for fuel by check No. 6 to Rogers Coal Co., 12.00.
Paid clerk's salary, check No. 7, 10.00.
Paid for labor, check No. 8, 16.50.
—9th—
Sold to Watkins Fish, 64 Prairie Av., 5 chests Oolong tea, 207# at .52.
Bought from Western Grocer Co., Chicago, 50 chests Oolong tea, 418#
at .39; 20 sacks Rio coffee, 876# at .22¼; 10 sacks Java coffee, 434#
at .25; 15 sacks Mocha coffee, 653# at .25; terms 30 days net, 2/10, f.
o. b. Omaha.
Received from Wright Noble cash in payment of our bill of Jan. 4th,
less 2% cash discount.
—10th—
Received from bank, check of Landis Snow protested for non-
payment, protest fees added 1.90.
Sent Laughlin Co. our check No. 9 in payment of account
—11th—
Sold to Raymond H. Moss, 182 Spring St., 5 chests Japan tea, 217# at
.48; 5 sacks Rio coffee, 214# at .28.
58. Sold to Watkins Fish, 10 sacks Mocha coffee, 424# at .32.
Cobb Willet paid their account in full, deducting 2% for cash.
—12th—
Deposited cash on hand.
Sold to Cobb Willet, 5 chests Japan tea, 213# at .48.
Sold for cash, 2 sacks Rio coffee, 88# at .28.
—13th—
Paid our note to Japan Importing Co., check No. 10.
Paid sundry office expenses from petty cash 3.60.
Sold to Wade Francis 3 sacks Rio coffee, 123# at .28; 2 sacks Mocha
coffee, 86# at .32.
Paid clerk's salary, ck. No. 11, 10.00.
Paid for labor, ck. No. 12, 16.50.
—15th—
Paid ½ month's salary to D. C. Hoadley, ck. No. 13, 75.00.
1st. Balance cash, first charging petty cash expenditures as a cash
payment.
2nd. Post purchase book, sales book, journal, and cash book.
3rd. Take a trial balance.
4th. Credit interest to partner's accounts.
5th. Take an inventory of stock on hand. The records show quantities
purchased and quantities sold. When the same goods have been
purchased at different prices, use the last price paid in figuring
inventory.
6th. Close accounts into trading and profit and loss accounts.
7th. Distribute net profits to partners' capital accounts.
59. COMBINED ORDER AND SALES RECORD
17. Instead of keeping separate order and sales books, both records may be
combined on one blank. This is accomplished by the use of order blanks
provided with columns for the distribution of sales to the different
departments. Before the order is filled these blanks are handled in the same
manner as those without distribution columns. When filled, the amounts are
extended in the proper columns and the invoice made. The orders are then
filed in a binder, each day's orders being kept together, and postings made
direct to customers' accounts. The footings are carried forward to the end of
the month and totals posted to sales accounts. The original orders thus
become a loose leaf sales book.
60. ABSTRACT OF SALES
18. When the order blank is used as a sales record, making a sales book
with a record of a single sale to a sheet, it is somewhat inconvenient to
determine from the footings the total sales for the day. This information is of
considerable value, as a knowledge of what is being done from day to day is
of importance to the principals of a business. Such a record is provided for
by an abstract of sales on a separate sheet. This abstract should show total
sales for each day, both cash and on account, divided by departments.
The blank may be made the same size as the order blanks, and filed in the
sales binder at the beginning of the month. Sales are recorded daily and
footings carried forward to the end of the month, when the totals may be
posted direct to the credit of the sales account in the general ledger. The
totals of sales on account will be posted to the debit of the sales controlling
account in the general ledger.
61. OUT FREIGHT
19. The proper treatment of freight paid on outgoing shipments is an
important question in accounting. When goods are sold at delivered prices,
the freight paid is one of the items of expense in selling the goods, and
when the books are closed the account will be closed into profit and loss.
However, in a wholesale business, freight is sometimes paid as an
accommodation to the customer when the goods have been sold at f. o. b.
prices. Although the amount should be added to the invoice it should not be
credited to the sales account as this would be taking credit for a fictitious
trading profit. Such an item should be made a special charge against the
customer by means of a journal entry.
Customer Dr.
Out Freight Cr.
65. SALES EXPENSE
20. In a wholesale or manufacturing business it is very desirable that the
exact cost of selling goods be known. Broadly, this cost is covered under the
general head of sales expense, but this is usually divided into several classes
of expenditures. The segregation of the various items of sales expense is
desirable for the purpose of determining the percentage of each. The items
which properly belong in sales expense depend somewhat on the nature of
the business. For example, traveling expenses are usually a direct sales
expense, but in some businesses they may be chargeable to the cost of
purchases. The items entering into sales expense of the average business
are: advertising, salaries of salesmen, traveling expenses of salesmen,
commissions paid on sales, cost of packing and shipping, out freight.
21. Advertising. This account should be charged with all expenditures for
publicity such as newspaper, magazine, street car, and bill board advertising,
cost of printing catalogs, booklets, and circulars. Where there is any reason
for so doing, the cost of the different classes of advertising can, of course,
be kept in separate accounts. The aim and object of advertising being to
increase the sale of goods, it is properly considered an item of sales
expense.
22. Salaries of Salesmen. This account is charged with all salaries paid to
salesmen whether traveling or working in the house. Commissions and
bonuses are sometimes included in this account, but it is usually considered
best to keep them in separate accounts.
23. Traveling Expenses of Salesmen. This account is charged with all
legitimate traveling expenses of salesmen, the specific items included
depending largely on the nature of the business. For example, in some
businesses a liberal allowance is made for the entertainment of customers,
while in others this item is never allowed. In any business the traveling
expense account requires careful scrutiny. Salesmen should be required to
furnish an itemized statement or voucher of expenses at stated intervals. For
convenience, this should be made on a form specially provided for the
purpose. One of the most convenient and popular expense vouchers is in the
form of a book of a convenient pocket size, with a page for each day of the
week and a summary of the week's expenses on the last page.
66. 24. Packing and Shipping. This account is charged with the entire cost of
packing goods for shipment. It includes such items as wages of shipping
clerk and his assistants, crates, lumber, boxes, and all other packing
materials.
67. TRIAL BALANCE BOOK
Traveler's Expense Book
25. To save rewriting the names of the accounts each month, a trial balance
book can be used to good advantage. These books are made to
68. accommodate six trial balances on a double page, and are sometimes made
with alternate short leaves so that twelve trial balances may be made with
one writing of the names. When the trial balance book is used, care must be
exercised in providing space for the addition of new accounts in each
section. Where separate sales and purchase ledgers are used, it is best to
provide a trial balance book for each ledger.
69. THE CHECK REGISTER
26. Large check books are cumbersome to handle and necessitating the
expenditure of much needless labor. Their use is rapidly giving way in
modern offices to the check register. The check register has several distinct
advantages. It exhibits, in compact form, a record of all checks issued and
can also be arranged to show deposits and balance in the bank. Distribution
columns can be provided with headings for the different expenditure
accounts, which makes of the check register a cash expenditure book. The
form should be varied to suit the business in which it is to be used. A typical
form is illustrated on page 37.
27. Checks in Pads. When the check register is used it is the usual custom
to have checks put up in pads. After the check is written, it is registered and
numbered to correspond to the register number. With the use of padded
checks, it is not necessary for the clerk who writes the check to know
anything about the bank balance.
Trial Balance Book
72. CASH RECEIVED BOOK
28. A cash book specially ruled for a record of cash received is used to
supplement the check register or cash expenditure book. Columns are
provided for the different classes of receipts, with one credit column. It is
assumed that all cash received is deposited, payments being made
exclusively by check. This does not refer to petty cash expenditures which
should be kept in a petty cash book or on envelope vouchers.
73. SAMPLE TRANSACTIONS
29. The following transactions illustrate the use of the special blanks and
books described.
D. A. Hall employs H. D. Snyder as traveling salesman for the purpose of
increasing his business, agreeing to pay him a salary of $150.00 per month
and expenses. He commences work on Feb. 11th. The amounts in the
ledgers stand as shown in the last model set illustrated and these
transactions are recorded:
—Feb. 11th.—
Paid Altman Sons
To balance account
Ck. No. 9 $350.00
—11th—
Paid Garson Co.
To balance account
Ck. No. 10 175.00
—11th—
Sold to Daniels Dean, Boone, Ia.
10 men's suits $7.50 $75.00
10 men's suits 6.75 67.50
20 boys' suits 2.00 40.00
182.50
Terms 2/10 N/30
—11th—
Sold to A. C. Petersen, Nevada, Ia.
10 men's overcoats 8.50 85.00
74. 10 men's suits 7.00 70.00
155.00
Terms 2/10 N/30
—11th—
Received from D. A. Marcus Son
Cash 139.65
Discount 2% 2.85
—11th—
Sold for cash
20 boys' suits 1.75 35.00
20 men's pants 2.00 40.00
—12th—
Received from John Gorham
Cash 132.30
Discount 2% 2.70
—12th—
Sold to Henry Cook, Iowa Falls, Ia.
5 men's suits 8.00 40.00
5 men's suits 5.75 28.75
5 boys' suits 2.00 10.00
5 boys' suits 1.75 8.75
87.50
Terms 2/10, N/30
—12th—
Sold to James Adams, Dennison, Ia.
15 men's suits 7.00 105.00
Terms 2/10, N/30
75. —12th—
Received from Geo. Golden
Cash $150.00
—13th—
Sold to D. A. Marcus Sons
10 men's pants 2.00 20.00
10 boys' suits 1.75 17.50
37.50
Terms 2/10, N/30
—13th—
Paid freight on shipment to
Henry Cook
Ck. No. 11 3.65
Charge to Cook
—13th—
Deposited cash 824.45
—13th—
Sold to S. H. Allen, Mason City, Ia.
30 men's pants 1.75 52.50
Terms 2/10, N/20
—14th—
Paid Adler Co.
Ck. No. 12 303.12
Discount 9.38
—15th—
Sold to Marx Sons, Charles City, Ia.
76. Welcome to our website – the perfect destination for book lovers and
knowledge seekers. We believe that every book holds a new world,
offering opportunities for learning, discovery, and personal growth.
That’s why we are dedicated to bringing you a diverse collection of
books, ranging from classic literature and specialized publications to
self-development guides and children's books.
More than just a book-buying platform, we strive to be a bridge
connecting you with timeless cultural and intellectual values. With an
elegant, user-friendly interface and a smart search system, you can
quickly find the books that best suit your interests. Additionally,
our special promotions and home delivery services help you save time
and fully enjoy the joy of reading.
Join us on a journey of knowledge exploration, passion nurturing, and
personal growth every day!
ebookbell.com